Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Oxid Med Cell Longev ; 2022: 1652244, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36299604

RESUMEN

Corilagin, a gallotannin, shows excellent antioxidant and anti-inflammatory effects. The NLRP3 inflammasome dysfunction has been implicated in a variety of inflammation diseases. However, it remains unclear how corilagin regulates the NLRP3 inflammasome to relieve gouty arthritis. In this study, bone marrow-derived macrophages (BMDMs) were pretreated with lipopolysaccharide (LPS) and then incubated with NLRP3 inflammasome agonists, such as adenine nucleoside triphosphate (ATP), nigericin, and monosodium urate (MSU) crystals. The MSU crystals were intra-articular injected to induce acute gouty arthritis. Here we showed that corilagin reduced lactate dehydrogenase (LDH) secretion and the proportion of propidium iodide- (PI-)stained cells. Corilagin suppressed the expression of N-terminal of the pyroptosis executive protein gasdermin D (GSDMD-NT). Corilagin restricted caspase-1 p20 and interleukin (IL)-1ß release. Meanwhile, corilagin attenuated ASC oligomerization and speck formation. Our findings confirmed that corilagin diminished NLRP3 inflammasome activation and macrophage pyroptosis. We further discovered that corilagin limited the mitochondrial reactive oxygen species (ROS) production and prevented the interaction between TXNIP and NLRP3, but ROS activator imiquimod could antagonize the inhibitory function of corilagin on NLRP3 inflammasome and macrophage pyroptosis. Additionally, corilagin ameliorated MSU crystals induced joint swelling, inhibited IL-1ß production, and abated macrophage and neutrophil migration into the joint capsule. Collectively, these results demonstrated that corilagin suppressed the ROS/TXNIP/NLRP3 pathway to repress inflammasome activation and pyroptosis and suggest its potential antioxidative role in alleviating NLRP3-dependent gouty arthritis.


Asunto(s)
Artritis Gotosa , Piroptosis , Humanos , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Taninos Hidrolizables/farmacología , Taninos Hidrolizables/uso terapéutico , Lipopolisacáridos/farmacología , Artritis Gotosa/tratamiento farmacológico , Artritis Gotosa/metabolismo , Ácido Úrico/uso terapéutico , Antioxidantes/farmacología , Nigericina/farmacología , Nigericina/uso terapéutico , Imiquimod/farmacología , Imiquimod/uso terapéutico , Propidio/farmacología , Propidio/uso terapéutico , Nucleósidos/farmacología , Caspasa 1/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Antiinflamatorios/farmacología , Adenosina Trifosfato/farmacología , Adenina/farmacología , Lactato Deshidrogenasas
2.
Curr Eye Res ; 47(10): 1366-1373, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35759617

RESUMEN

PURPOSE: The purpose of this research was to explore the antifungal and anti-inflammatory effects of perillaldehyde (PAE) in Aspergillus fumigatus (A. fumigatus) keratitis and the underlying mechanism. METHODS: The biofilm formation, adherence assay, and propidium iodide uptake test were used to determine the possible mechanism of PAE in terms of antifungal effects in vitro. The severity of corneal infection was evaluated by clinical scores. The immunofluorescence staining (IFS) was adopted to detect the number of macrophages in infected corneas. Draize test was performed to assess the ocular toxicity of PAE. Real-time polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and Western blot reflected the expression of inflammatory cytokines and Lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) in mice corneas and RAW264.7 cells. RESULTS: PAE was able to inhibit the formation of biofilm, reduce conidial adhesion, and damage the integrity of membranes to exert antifungal activity. In C57BL/6 mice models, PAE alleviated the severity of infected corneas, reduced the recruitment of macrophages and had low ocular toxicity. In addition, the mRNA and protein levels of TNF-α, CCL-2, and LOX-1 could be significantly decreased by the application of PAE after A. fumigatus infection in vivo and in vitro. CONCLUSION: Our study indicated that PAE protected against A. fumigatus keratitis by reducing fungal load, accumulation of macrophages, and inhibiting the expression of inflammatory cytokines.


Asunto(s)
Infecciones Fúngicas del Ojo , Queratitis , Animales , Antiinflamatorios , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Aspergillus fumigatus/metabolismo , Citocinas/genética , Modelos Animales de Enfermedad , Infecciones Fúngicas del Ojo/microbiología , Queratitis/tratamiento farmacológico , Queratitis/metabolismo , Queratitis/prevención & control , Ratones , Ratones Endogámicos C57BL , Monoterpenos , Propidio/uso terapéutico , ARN Mensajero , Receptores Depuradores de Clase E/genética , Receptores Depuradores de Clase E/metabolismo , Receptores Depuradores de Clase E/uso terapéutico , Neuropatía Óptica Tóxica , Factor de Necrosis Tumoral alfa
3.
Biochimie ; 201: 7-17, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35764196

RESUMEN

Non-small cell lung cancer (NSCLC) is a major cause of global cancer mortalities and accounts for approximately 80-85% of reported lung cancer cases. Conventional chemotherapeutics show limited application because of poor tumor selectivity and acquired drug resistance. Antimicrobial peptides (AMPs) have gained much attention as potential anticancer therapeutics owing to their high potency and high target selectivity and specificity with limited scope for drug resistance. In this study, D-LAK (D-LAK-120A), a cationic AMP, was evaluated for its anticancer efficacy in various NSCLC cell lines. D-LAK peptide demonstrated enhanced cytotoxicity in A549, H358, H1975, and HCC827 cell lines with inhibitory concentrations between 4.0 and 5.5 µM. An increase in the lactate dehydrogenase (LDH) levels and propidium iodide (PI) uptake across compromised membrane suggested membranolytic activity as an inhibition pathway. In addition, we found D-LAK induced lung cancer cell apoptosis and arrested cells in the S phase (DNA synthesis) of cell cycle. Moreover, a decreased mitochondrial membrane potential and elevated ROS levels were observed after D-LAK treatment, suggesting induction of mitochondria-mediated apoptosis. Additionally, D-LAK inhibited single cell proliferation and cancer cell migration in vitro. The tumor reduction observed in the 3D spheroid assay further suggests the potential use of D-LAK as an anticancer agent for NSCLC treatment. Our results postulate innovative insights on the anticancer mechanism of D-LAK, which may contribute to its further development into preclinical studies and a potential therapeutic.


Asunto(s)
Antineoplásicos , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Péptidos Antimicrobianos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular , ADN , Lactato Deshidrogenasas , Neoplasias Pulmonares/genética , Propidio/farmacología , Propidio/uso terapéutico , Especies Reactivas de Oxígeno
4.
Anticancer Agents Med Chem ; 22(18): 3114-3124, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35473535

RESUMEN

BACKGROUND: Lung tumors express high levels of aromatase enzyme compared to surrounding normal tissue. Inhibition of aromatase has emerged as a recent therapeutic approach for the treatment of breast cancer. However, the role of aromatase inhibition in lung cancer treatment requires further investigation. METHODS: The anti-proliferative effects of aromatase inhibitors were evaluated by MTT assay. Cell migration was assessed using a wound healing assay. The mechanism of cell death was determined using the annexin VFITC/ propidium iodide staining flow cytometry method. The soft agar colony formation assay evaluated cells' capability to form colonies. RESULT: Exemestane and curcumin significantly inhibited the growth of lung cancer cell lines in a dose- and timedependent manner. The IC50 values after 48 hours of treatment with exemestane were 176, 180, and 120 µM in A549, H661, and H1299, respectively. Curcumin IC50 values after 48 hours were 80, 43, and 68 µM in A549, H661, and H1299, respectively. The combined treatment of exemestane or curcumin with cisplatin, raloxifene, and celecoxib resulted in a synergistic effect in the A549 lung cell line with a combination index of less than 1, suggesting synergism. Exemestane resulted in approximately 96% inhibition of wound closure at 100 µM, while curcumin resulted in approximately 63% inhibition of wound closure at 50 µM. Exemestane and curcumin inhibited the formation of cell colonies by reducing the number and size of formed colonies of A549, H661, and H1299 cell lines in a concentration dependent manner. Exemestane and curcumin had significantly induced apoptosis in A549 cells compared to control of untreated cells. CONCLUSION: Aromatase inhibition by exemestane or curcumin had significantly inhibited the growth of lung cancer cell lines, synergized with cisplatin, raloxifene, and celecoxib, suppressed lung cancer cell migratory potential, induced apoptosis, and reduced colony formation of lung cancer cells.


Asunto(s)
Curcumina , Neoplasias Pulmonares , Agar/farmacología , Agar/uso terapéutico , Anexinas/farmacología , Anexinas/uso terapéutico , Apoptosis , Aromatasa/metabolismo , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/uso terapéutico , Celecoxib/farmacología , Línea Celular Tumoral , Proliferación Celular , Cisplatino/farmacología , Curcumina/farmacología , Curcumina/uso terapéutico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Propidio/farmacología , Propidio/uso terapéutico , Clorhidrato de Raloxifeno/uso terapéutico
5.
J Biol Chem ; 294(49): 18624-18638, 2019 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-31649034

RESUMEN

Repeat-associated non-AUG (RAN) translation is a noncanonical translation initiation event that occurs at nucleotide-repeat expansion mutations that are associated with several neurodegenerative diseases, including fragile X-associated tremor ataxia syndrome (FXTAS), ALS, and frontotemporal dementia (FTD). Translation of expanded repeats produces toxic proteins that accumulate in human brains and contribute to disease pathogenesis. Consequently, RAN translation constitutes a potentially important therapeutic target for managing multiple neurodegenerative disorders. Here, we adapted a previously developed RAN translation assay to a high-throughput format to screen 3,253 bioactive compounds for inhibition of RAN translation of expanded CGG repeats associated with FXTAS. We identified five diverse small molecules that dose-dependently inhibited CGG RAN translation, while relatively sparing canonical translation. All five compounds also inhibited RAN translation of expanded GGGGCC repeats associated with ALS and FTD. Using CD and native gel analyses, we found evidence that three of these compounds, BIX01294, CP-31398, and propidium iodide, bind directly to the repeat RNAs. These findings provide proof-of-principle supporting the development of selective small-molecule RAN translation inhibitors that act across multiple disease-causing repeats.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Ataxia/genética , Síndrome del Cromosoma X Frágil/genética , Temblor/genética , Expansión de Repetición de Trinucleótido/genética , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Animales , Ataxia/tratamiento farmacológico , Azepinas/farmacología , Azepinas/uso terapéutico , Células Cultivadas , Dicroismo Circular , Expansión de las Repeticiones de ADN/efectos de los fármacos , Expansión de las Repeticiones de ADN/genética , Evaluación Preclínica de Medicamentos , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Células HEK293 , Humanos , Enfermedades Neurodegenerativas/genética , Propidio/farmacología , Propidio/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Ratas , Temblor/tratamiento farmacológico , Expansión de Repetición de Trinucleótido/efectos de los fármacos
6.
ISME J ; 7(4): 697-706, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23190732

RESUMEN

High-throughput pyrosequencing and quantitative PCR (Q-PCR) analysis offer greatly improved accuracy and depth of characterisation of lower respiratory infections. However, such approaches suffer from an inability to distinguish between DNA derived from viable and non-viable bacteria. This discrimination represents an important step in characterising microbial communities, particularly in contexts with poor clearance of material or high antimicrobial stress, as non-viable bacteria and extracellular DNA can contribute significantly to analyses. Pre-treatment of samples with propidium monoazide (PMA) is an effective approach to non-viable cell exclusion (NVCE). However, the impact of NVCE on microbial community characteristics (abundance, diversity, composition and structure) is not known. Here, adult cystic fibrosis (CF) sputum samples were used as a paradigm. The effects of PMA treatment on CF sputum bacterial community characteristics, as analysed by pyrosequencing and enumeration by species-specific (Pseudomonas aeruginosa) and total bacterial Q-PCR, were assessed. At the local community level, abundances of both total bacteria and of P. aeruginosa were significantly lower in PMA-treated sample portions. Meta-analysis indicated no overall significant differences in diversity; however, PMA treatment resulted in a significant alteration in local community membership in all cases. In contrast, at the metacommunity level, PMA treatment resulted in an increase in community evenness, driven by an increase in diversity, predominately representing rare community members. Importantly, PMA treatment facilitated the detection of both recognised and emerging CF pathogens, significantly influencing 'core' and 'satellite' taxa group membership. Our findings suggest failure to implement NVCE may result in skewed bacterial community analyses.


Asunto(s)
Azidas/uso terapéutico , Bacterias/clasificación , Bacterias/efectos de los fármacos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/microbiología , Propidio/análogos & derivados , Infecciones del Sistema Respiratorio/microbiología , Adulto , Bacterias/genética , Bacterias/aislamiento & purificación , Fibrosis Quística/complicaciones , Humanos , Propidio/uso terapéutico , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Esputo/microbiología
7.
Anal Chem ; 81(9): 3517-22, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19344145

RESUMEN

A microfabricated cell-based testing device for electrochemotherapy (ECT) has been developed; it mimics a clinical electroporator with a circular needle array and maintains a similar electric field strength distribution. Until now, the performance between electroporators having two- and six-needle circular array electrodes, which are the general needle-type clinical electroporators for ECT, has not been evaluated systemically, although many studies have investigated the efficacy of ECT on cancer cells. In this study, the cell-based performance on the newly developed ECT testing device was analyzed in two- and six-electrode modes using propidium iodide and bleomycin, and the electroporation characteristics were characterized. With on-chip fluorescence-based assays, the proposed device offers time- and cost-effective experimental procedures compared with conventional assays, and the results can reflect the electric characteristics of clinical electroporators.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Carcinoma Ductal/tratamiento farmacológico , Carcinoma Ductal/patología , Electroquimioterapia , Microtecnología/instrumentación , Bleomicina/metabolismo , Bleomicina/farmacología , Bleomicina/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Electrodos , Fluorescencia , Humanos , Propidio/metabolismo , Propidio/uso terapéutico
8.
Curr Med Chem ; 14(25): 2654-79, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17979717

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the central nervous system (CNS) which is the most common cause of dementia in the elderly. It is characterized by the deficits in the cholinergic system and presence of characteristic hallmarks: neurofibrillary tangles and amyloid plaques. Since the cholinergic system plays an important role in the regulation of learning and memory processes it became a target for the design of anti-alzheimer drugs. Cholinesterase inhibitors enhance cholinergic transmission indirectly, by inhibiting the enzyme which hydrolyses acetylcholine. It has been also demonstrated that acetylcholinesterase (AChE) is involved in the development of amyloid plaques. Therefore, substances which are AChE inhibitors (AChEI) are the only drugs approved for the symptomatic treatment of AD. This review presents the main classes of cholinesterase inhibitors developed recently for the treatment of AD. We have started with the analogues of the existing drugs: tacrine, donepezil, rivastigmine and galantamine which are still of interest for many research groups. Among them there is a very interesting group--dual binding site inhibitors characterized by increased inhibitory potency against AChE and amyloid plaques formation. There is also a group of compounds with additional properties such as: antioxidant activity, affinity to 5-HT(3) receptors, inhibition of N-methyltransferase that metabolize histamine, which can be beneficial for the treatment of AD. Furthermore there are some interesting compounds which belong to different chemical groups also of natural origin. In this review we sum up current research concerned with development of AChEIs which can be more effective in the future treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Inhibidores de la Colinesterasa/uso terapéutico , Alcaloides/uso terapéutico , Sitios de Unión , Donepezilo , Galantamina/análogos & derivados , Galantamina/uso terapéutico , Humanos , Indanos/uso terapéutico , Alcaloides Indólicos/uso terapéutico , Isoquinolinas/uso terapéutico , Fenilcarbamatos/uso terapéutico , Fisostigmina/análogos & derivados , Fisostigmina/uso terapéutico , Piperidinas/uso terapéutico , Propidio/análogos & derivados , Propidio/uso terapéutico , Rivastigmina , Esteroides/uso terapéutico , Tacrina/análogos & derivados , Tacrina/uso terapéutico
9.
Farmaco ; 60(6-7): 465-73, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15878569

RESUMEN

Notwithstanding the criticism to the so called " cholinergic hypothesis", the therapeutic strategies for the treatment of Alzheimer's disease (AD) have been mainly centered on the restoration of cholinergic functionality and, until the last year, the only drugs licensed for the management of AD were the acetycholinesterase (AChE) inhibitors. Target enzyme AChE consists of a narrow gorge with two separate ligand binding sites: an acylation site at the bottom of the gorge containing the catalytic triad and a peripheral site located at the gorge rim, which encompasses binding sites for allosteric ligands. The aim of this short review is to update the knowledge on heterocyclic AChE inhibitors able to interact with the two sites of enzymes, structurally related to the well known inhibitors physostigmine, rivastigmine and propidium. The therapeutic potential of the dual site inhibithors in inhibiting amyloid-beta aggregatrion and deposition is also briefly summarised.


Asunto(s)
Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/uso terapéutico , Compuestos Heterocíclicos/uso terapéutico , Acetilcolinesterasa/química , Acilación , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Sitios de Unión , Inhibidores de la Colinesterasa/química , Compuestos Heterocíclicos/química , Humanos , Estructura Molecular , Fenilcarbamatos/química , Fenilcarbamatos/uso terapéutico , Fisostigmina/química , Fisostigmina/uso terapéutico , Propidio/química , Propidio/uso terapéutico , Rivastigmina
10.
Clin Cancer Res ; 9(8): 3183-9, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12912971

RESUMEN

PURPOSE: Because survival for patients with oral cancer has not improved over the past 25 years, new approaches for treatment are needed. Targeted molecular therapy against epidermal growth factor receptor (EGFR) has shown promise as an adjuvant therapy in preliminary studies in several solid tumors, including head and neck cancer. The objective of this study was to determine the efficacy of paclitaxel and PKI166, a novel inhibitor of EGFR, against oral cavity cancer. EXPERIMENTAL DESIGN AND RESULTS: JMAR human oral cancer cells were pretreated for 1 h with PKI166 and then stimulated with epidermal growth factor. EGFR-specific tyrosine kinase autophosphorylation measured by Western immunoblotting was inhibited by PKI166 in a dose-dependent fashion at all doses tested (0.01-1 micro M). Next, the induction of apoptosis in JMAR cells treated with paclitaxel (0.001 to 0.1 micro M) with or without PKI166 (0, 1, or 2 micro M) was determined using a propidium iodide assay. The addition of 2.0 micro M PKI166 significantly increased tumor cell death, shifting the amount of paclitaxel needed to induce apoptosis in 50% of cells from 0.1 to 0.001 micro M. These in vitro findings were confirmed using an orthotopic model of oral cancer. JMAR oral cancer cells were implanted into the tongues of nude mice. After lingual tumors developed, mice were randomized into four groups (n = 10): (a) oral PKI166 (100 mg/kg); (b) i.p. paclitaxel (200 micro g/wk); (c) PKI166 and paclitaxel; or (d) placebo. Mice treated with PKI166/paclitaxel demonstrated a significant increase in survival (P = 0.028). After necropsy, all tongue tumors were evaluated for apoptosis by the terminal deoxynucleotidyl transferase-mediated nick end labeling assay. A greater apoptotic fraction of tumor cells was found in tumors of mice treated with paclitaxel and PKI166 as compared with the other treatment groups (136.4 versus 37.8; P = 0.016). CONCLUSIONS: Combination therapy with paclitaxel and PKI166 prolongs survival in an orthotopic preclinical model of tongue cancer by increasing programmed cell death of oral cancer.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis , Receptores ErbB/antagonistas & inhibidores , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/mortalidad , Paclitaxel/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Animales , Western Blotting , Muerte Celular , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Neoplasias de Cabeza y Cuello/patología , Humanos , Etiquetado Corte-Fin in Situ , Ligandos , Masculino , Ratones , Ratones Desnudos , Microscopía Fluorescente , Neoplasias de la Boca/metabolismo , Neoplasias/patología , Fosforilación , Propidio/uso terapéutico , Lengua/patología , Neoplasias de la Lengua/tratamiento farmacológico , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA