Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Molecules ; 26(4)2021 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-33672721

RESUMEN

The ongoing coronavirus pandemic has been a burden on the worldwide population, with mass fatalities and devastating socioeconomic consequences. It has particularly drawn attention to the lack of approved small-molecule drugs to inhibit SARS coronaviruses. Importantly, lessons learned from the SARS outbreak of 2002-2004, caused by severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1), can be applied to current drug discovery ventures. SARS-CoV-1 and SARS-CoV-2 both possess two cysteine proteases, the main protease (Mpro) and the papain-like protease (PLpro), which play a significant role in facilitating viral replication, and are important drug targets. The non-covalent inhibitor, GRL-0617, which was found to inhibit replication of SARS-CoV-1, and more recently SARS-CoV-2, is the only PLpro inhibitor co-crystallised with the recently solved SARS-CoV-2 PLpro crystal structure. Therefore, the GRL-0617 structural template and pharmacophore features are instrumental in the design and development of more potent PLpro inhibitors. In this work, we conducted scaffold hopping using GRL-0617 as a reference to screen over 339,000 ligands in the chemical space using the ChemDiv, MayBridge, and Enamine screening libraries. Twenty-four distinct scaffolds with structural and electrostatic similarity to GRL-0617 were obtained. These proceeded to molecular docking against PLpro using the AutoDock tools. Of two compounds that showed the most favourable predicted binding affinities to the target site, as well as comparable protein-ligand interactions to GRL-0617, one was chosen for further analogue-based work. Twenty-seven analogues of this compound were further docked against the PLpro, which resulted in two additional hits with promising docking profiles. Our in silico pipeline consisted of an integrative four-step approach: (1) ligand-based virtual screening (scaffold-hopping), (2) molecular docking, (3) an analogue search, and, (4) evaluation of scaffold drug-likeness, to identify promising scaffolds and eliminate those with undesirable properties. Overall, we present four novel, and lipophilic, scaffolds obtained from an exhaustive search of diverse and uncharted regions of chemical space, which may be further explored in vitro through structure-activity relationship (SAR) studies in the search for more potent inhibitors. Furthermore, these scaffolds were predicted to have fewer off-target interactions than GRL-0617. Lastly, to our knowledge, this work contains the largest ligand-based virtual screen performed against GRL-0617.


Asunto(s)
Antivirales/química , COVID-19/enzimología , Proteasas 3C de Coronavirus , Inhibidores de Cisteína Proteinasa/química , Simulación del Acoplamiento Molecular , SARS-CoV-2/enzimología , Antivirales/uso terapéutico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/uso terapéutico , Evaluación Preclínica de Medicamentos , Humanos , Tratamiento Farmacológico de COVID-19
2.
Molecules ; 26(4)2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33669720

RESUMEN

Coronavirus desease 2019 (COVID-19) is responsible for more than 1.80 M deaths worldwide. A Quantitative Structure-Activity Relationships (QSAR) model is developed based on experimental pIC50 values reported for a structurally diverse dataset. A robust model with only five descriptors is found, with values of R2 = 0.897, Q2LOO = 0.854, and Q2ext = 0.876 and complying with all the parameters established in the validation Tropsha's test. The analysis of the applicability domain (AD) reveals coverage of about 90% for the external test set. Docking and molecular dynamic analysis are performed on the three most relevant biological targets for SARS-CoV-2: main protease, papain-like protease, and RNA-dependent RNA polymerase. A screening of the DrugBank database is executed, predicting the pIC50 value of 6664 drugs, which are IN the AD of the model (coverage = 79%). Fifty-seven possible potent anti-COVID-19 candidates with pIC50 values > 6.6 are identified, and based on a pharmacophore modelling analysis, four compounds of this set can be suggested as potent candidates to be potential inhibitors of SARS-CoV-2. Finally, the biological activity of the compounds was related to the frontier molecular orbitals shapes.


Asunto(s)
Antivirales/química , COVID-19/enzimología , Proteasas 3C de Coronavirus , Inhibidores de Cisteína Proteinasa/química , Bases de Datos de Compuestos Químicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , ARN Polimerasa Dependiente del ARN , SARS-CoV-2/enzimología , Antivirales/uso terapéutico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Inhibidores de Cisteína Proteinasa/uso terapéutico , Evaluación Preclínica de Medicamentos , Relación Estructura-Actividad Cuantitativa , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/química , Tratamiento Farmacológico de COVID-19
3.
Molecules ; 26(5)2021 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-33668085

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has emerged to be the greatest threat to humanity in the modern world and has claimed nearly 2.2 million lives worldwide. The United States alone accounts for more than one fourth of 100 million COVID-19 cases across the globe. Although vaccination against SARS-CoV-2 has begun, its efficacy in preventing a new or repeat COVID-19 infection in immunized individuals is yet to be determined. Calls for repurposing of existing, approved, drugs that target the inflammatory condition in COVID-19 are growing. Our initial gene ontology analysis predicts a similarity between SARS-CoV-2 induced inflammatory and immune dysregulation and the pathophysiology of rheumatoid arthritis. Interestingly, many of the drugs related to rheumatoid arthritis have been found to be lifesaving and contribute to lower COVID-19 morbidity. We also performed in silico investigation of binding of epigallocatechin gallate (EGCG), a well-known catechin, and other catechins on viral proteins and identified papain-like protease protein (PLPro) as a binding partner. Catechins bind to the S1 ubiquitin-binding site of PLPro, which might inhibit its protease function and abrogate SARS-CoV-2 inhibitory function on ubiquitin proteasome system and interferon stimulated gene system. In the realms of addressing inflammation and how to effectively target SARS-CoV-2 mediated respiratory distress syndrome, we review in this article the available knowledge on the strategic placement of EGCG in curbing inflammatory signals and how it may serve as a broad spectrum therapeutic in asymptomatic and symptomatic COVID-19 patients.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Catequina/análogos & derivados , Proteasas 3C de Coronavirus , Inhibidores de Cisteína Proteinasa , SARS-CoV-2/enzimología , Té/química , Antivirales/química , Antivirales/uso terapéutico , Sitios de Unión , COVID-19/enzimología , COVID-19/epidemiología , Catequina/química , Catequina/uso terapéutico , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/uso terapéutico , Humanos
4.
Cell Commun Signal ; 19(1): 24, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33627137

RESUMEN

BACKGROUND: The oncogenic transcript factor c-Maf is stabilized by the deubiquitinase Otub1 and promotes myeloma cell proliferation and confers to chemoresistance. Inhibition of the Otub1/c-Maf axis is a promising therapeutic target, but there are no inhibitors reported on this specific axis. METHODS: A luciferase assay was applied to screen potential inhibitors of Otub1/c-Maf. Annexin V staining/flow cytometry was applied to evaluate cell apoptosis. Immunoprecipitation was applied to examine protein ubiquitination and interaction. Xenograft models in nude mice were used to evaluate anti-myeloma activity of AVT. RESULTS: Acevaltrate (AVT), isolated from Valeriana glechomifolia, was identified based on a bioactive screen against the Otub1/c-Maf/luciferase system. AVT disrupts the interaction of Otub1/c-Maf thus inhibiting Otub1 activity and leading to c-Maf polyubiquitination and subsequent degradation in proteasomes. Consistently, AVT inhibits c-Maf transcriptional activity and downregulates the expression of its target genes key for myeloma growth and survival. Moreover, AVT displays potent anti-myeloma activity by triggering myeloma cell apoptosis in vitro and impairing myeloma xenograft growth in vivo but presents no marked toxicity. CONCLUSIONS: The natural product AVT inhibits the Otub1/c-Maf axis and displays potent anti-myeloma activity. Given its great safety and efficacy, AVT could be further developed for MM treatment. Video Abstract.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Iridoides/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-maf/antagonistas & inhibidores , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cisteína Endopeptidasas/genética , Inhibidores de Cisteína Proteinasa/farmacología , Femenino , Humanos , Iridoides/farmacología , Ratones Endogámicos BALB C , Ratones Desnudos , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/metabolismo
5.
J Cell Physiol ; 234(2): 1001-1007, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30145792

RESUMEN

Dysregulation of neuronal Ca2+ and oxidative stress plays an important role in the activation of cysteine proteases including calpains and caspases that contribute to neuronal death. In neurodegenerative diseases, traumatic brain injury, stroke, and neuropathic pain calpain activities are markedly increased. Melatonin is a beneficial supplement in the treatment of central nervous system (CNS) disorders. Melatonin is a potent antioxidant and works as a free-radical scavenger to regulate a large number of molecular pathways, including oxidative stress, inflammation, apoptosis, and cell death under different pathological conditions. However, limited studies have evaluated the inhibitory effect of melatonin on calpains. This review summarizes the current knowledge related to the effects of melatonin on calpains in some of the common CNS disorders.


Asunto(s)
Calpaína/antagonistas & inhibidores , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Sistema Nervioso Central/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/uso terapéutico , Melatonina/uso terapéutico , Animales , Calpaína/metabolismo , Sistema Nervioso Central/enzimología , Sistema Nervioso Central/patología , Sistema Nervioso Central/fisiopatología , Enfermedades del Sistema Nervioso Central/enzimología , Enfermedades del Sistema Nervioso Central/patología , Enfermedades del Sistema Nervioso Central/fisiopatología , Humanos , Transducción de Señal
6.
Chem Biol Drug Des ; 92(3): 1585-1596, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29729080

RESUMEN

Chemotherapy is currently the only effective approach to treat all forms of leishmaniasis. However, its effectiveness is severely limited due to high toxicity, long treatment length, drug resistance, or inadequate mode of administration. As a consequence, there is a need to identify new molecular scaffolds and targets as potential therapeutics for the treatment of this disease. We report a small series of 1,2-substituted-1H-benzo[d]imidazole derivatives (9a-d) showing affinity in the submicromolar range (Ki  = 0.15-0.69 µM) toward Leishmania mexicanaCPB2.8ΔCTE, one of the more promising targets for antileishmanial drug design. The compounds confirmed activity in vitro against intracellular amastigotes of Leishmania infantum with the best result being obtained with derivative 9d (IC50  = 6.8 µM), although with some degree of cytotoxicity (CC50  = 8.0 µM on PMM and CC50  = 32.0 µM on MCR-5). In silico molecular docking studies and ADME-Tox properties prediction were performed to validate the hypothesis of the interaction with the intended target and to assess the drug-likeness of these derivatives.


Asunto(s)
Bencimidazoles/química , Proteasas de Cisteína/metabolismo , Inhibidores de Cisteína Proteinasa/química , Leishmania mexicana/enzimología , Proteínas Protozoarias/antagonistas & inhibidores , Antiprotozoarios/síntesis química , Antiprotozoarios/metabolismo , Antiprotozoarios/uso terapéutico , Antiprotozoarios/toxicidad , Bencimidazoles/metabolismo , Bencimidazoles/uso terapéutico , Bencimidazoles/toxicidad , Sitios de Unión , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteasas de Cisteína/química , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/uso terapéutico , Inhibidores de Cisteína Proteinasa/toxicidad , Evaluación Preclínica de Medicamentos , Pruebas de Enzimas , Humanos , Enlace de Hidrógeno , Concentración 50 Inhibidora , Leishmaniasis/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Estructura Terciaria de Proteína , Proteínas Protozoarias/metabolismo
7.
Int J Biol Macromol ; 95: 734-742, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27916569

RESUMEN

Phytocystatins are cysteine proteinase inhibitors present in plants. They play crucial role in maintaining protease-anti protease balance and are involved in various endogenous processes. Thus, they are suitable and convenient targets for genetic engineering which makes their isolation and characterisation from different sources the need of the hour. In the present study a phytocystatin has been isolated from garlic (Allium sativum) by a simple two-step process using ammonium sulphate fractionation and gel filtration chromatography on Sephacryl S-100HR with a fold purification of 152.6 and yield 48.9%. A single band on native gel electrophoresis confirms the homogeneity of the purified inhibitor. The molecular weight of the purified inhibitor was found to be 12.5kDa as determined by SDS-PAGE and gel filtration chromatography. The garlic phytocystatin was found to be stable under broad range of pH (6-8) and temperature (30°C-60°C). Kinetic studies suggests that garlic phytocystatins are reversible and non-competitive inhibitors having highest affinity for papain followed by ficin and bromelain. UV and fluorescence spectroscopy revealed significant conformational change upon garlic phytocystatin-papain complex formation. Secondary structure analysis was performed using CD and FTIR. Garlic phytocystatin possesses 33.9% alpha-helical content as assessed by CD spectroscopy.


Asunto(s)
Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/metabolismo , Ajo , Animales , Carbohidratos/análisis , Enfermedades Cardiovasculares/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/inmunología , Inhibidores de Cisteína Proteinasa/uso terapéutico , Concentración de Iones de Hidrógeno , Cinética , Peso Molecular , Estabilidad Proteica , Estructura Secundaria de Proteína , Análisis Espectral , Compuestos de Sulfhidrilo/análisis , Temperatura
8.
Mini Rev Med Chem ; 16(17): 1374-1391, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27156518

RESUMEN

Human African Trypanosomiasis (HAT) is an endemic parasitic disease of sub-Saharan Africa, caused by two subspecies of protozoa belonging to Trypanosoma genus: T. brucei gambiense and T. brucei rhodesiense. In this context the inhibition of the papain-family cysteine proteases rhodesain and TbCatB has to be considered a promising strategy for HAT treatment. Rhodesain, the major cathepsin L-like cysteine protease of T. brucei rhodesiense, is a lysosomal protease essential for parasite survival. It is involved in parasite invasivity, allowing it to cross the blood-brain barrier (BBB) of the human host, causing the second lethal stage of the disease. Moreover, it plays an important role in immunoevasion, being involved in the turnover of variant surface glycoproteins of the T. brucei coat and in the degradation of immunoglobulins, avoiding a specific immune response by the host cells. On the other hand TbCatB, a cathepsin B-like cysteine protease, present in minor abundance in T. brucei, showed a key role in the degradation of host transferrin, which is necessary for iron acquisition by the parasite. In this review article we now discuss the most active peptide, peptidomimetic and non-peptide rhodesain and TbCatB inhibitors as valuable strategy to treat HAT, due also to the complementary role of the two T. brucei proteases.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/metabolismo , Proteínas Protozoarias/metabolismo , Tripanocidas/metabolismo , Aziridinas/química , Aziridinas/metabolismo , Aziridinas/farmacología , Aziridinas/uso terapéutico , Barrera Hematoencefálica/metabolismo , Cisteína Endopeptidasas/química , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores de Cisteína Proteinasa/uso terapéutico , Humanos , Proteínas Protozoarias/antagonistas & inhibidores , Sulfonas/química , Sulfonas/metabolismo , Sulfonas/farmacología , Sulfonas/uso terapéutico , Tripanocidas/química , Tripanocidas/farmacología , Tripanocidas/uso terapéutico , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/enzimología , Tripanosomiasis Africana/tratamiento farmacológico
9.
Physiol Res ; 65(3): 537-41, 2016 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-27070748

RESUMEN

Several diseases induce hypermetabolism, which is characterized by increases in resting energy expenditures (REE) and whole body protein loss. Exaggerated protein degradation is thought to be the driving force underlying this response. The effects of caspase and calpain inhibitors on REE in physiological and hypermetabolic conditions, however, are unknown. Thus, we studied whether MDL28170 (calpain inhibitor) or z-VAD-fmk (caspase inhibitor) affect REE under physiological conditions and during hypermetabolism post-burn. Rats were treated five times weekly and observed for 6 weeks. Treatment was started 2 h (early) or 48 h (late) after burn. In normal rats, MDL28170 transiently increased REE to 130 % of normal during week 2-4. z-VAD-fmk reduced REE by 20-25 % throughout the observation period. Within 14 days after burns, REE increased to 130+/-5 %. Whereas MDL28170/early treatment did not affect REE, MDL28170/late transiently increased REE to 180+/-10 % of normal by week 4 post-burn. In contrast, with z-VAD-fmk/early REE remained between 90-110 % of normal post-burn. z-VAD-fmk/late did not affect burn-induced increases in REE. These data suggest that caspase cascades contribute to the development of hypermetabolism and that burn-induced hypermetabolism can be pharmacologically modulated. Our data point towards caspase cascades as possible therapeutic targets to attenuate hypermetabolism after burns, and possibly in other catabolic disease processes.


Asunto(s)
Clorometilcetonas de Aminoácidos/uso terapéutico , Inhibidores de Caspasas/uso terapéutico , Inhibidores de Cisteína Proteinasa/uso terapéutico , Dipéptidos/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Enfermedades Metabólicas/tratamiento farmacológico , Clorometilcetonas de Aminoácidos/farmacología , Animales , Quemaduras/complicaciones , Inhibidores de Caspasas/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Evaluación Preclínica de Medicamentos , Masculino , Enfermedades Metabólicas/etiología , Proyectos Piloto , Ratas Sprague-Dawley
10.
Biol Pharm Bull ; 37(6): 1021-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24882413

RESUMEN

Tribulus terrestris fruits are well known for their usage in pharmaceutical preparations and food supplements. The methanol extract of T. terrestris fruits showed potent inhibition against the papain-like protease (PLpro), an essential proteolylic enzyme for protection to pathogenic virus and bacteria. Subsequent bioactivity-guided fractionation of this extract led to six cinnamic amides (1-6) and ferulic acid (7). Compound 6 emerged as new compound possessing the very rare carbinolamide motif. These compounds (1-7) were evaluated for severe acute respiratory syndrome coronavirus (SARS-CoV) PLpro inhibitory activity to identify their potencies and kinetic behavior. Compounds (1-6) displayed significant inhibitory activity with IC50 values in the range 15.8-70.1 µM. The new cinnamic amide 6 was found to be most potent inhibitor with an IC50 of 15.8 µM. In kinetic studies, all inhibitors exhibited mixed type inhibition. Furthermore, the most active PLpro inhibitors (1-6) were proven to be present in the native fruits in high quantities by HPLC chromatogram and liquid chromatography with diode array detection and electrospray ionization mass spectrometry (LC-DAD-ESI/MS).


Asunto(s)
Cinamatos/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Extractos Vegetales/química , Tribulus/química , Proteínas Virales/antagonistas & inhibidores , Amidas , Cinamatos/aislamiento & purificación , Cinamatos/uso terapéutico , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/genética , Inhibidores de Cisteína Proteinasa/aislamiento & purificación , Inhibidores de Cisteína Proteinasa/uso terapéutico , Relación Dosis-Respuesta a Droga , Escherichia coli/genética , Frutas/química , Humanos , Concentración 50 Inhibidora , Cinética , Estructura Molecular , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología , Relación Estructura-Actividad , Proteínas Virales/genética
11.
FASEB J ; 28(8): 3564-78, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24776743

RESUMEN

The periodontal pathogen Porphyromonas gingivalis produces a unique class of cysteine proteinases termed gingipains that comprises Arg-gingipain (Rgp) and Lys-gingipain (Kgp). Growing evidence indicates that these 2 types of gingipains synergistically contribute to the entire virulence of the organism and increase the risk of periodontal disease (PD) by disrupting the host immune system and degrading the host tissue and plasma proteins. Therefore, a dual inhibitor of both gingipains would have attractive clinical potential for PD therapy. In this study, a novel, potent, dual inhibitor of Rgp and Kgp was developed through structure-based drug design, and its biological potency was evaluated in vitro and in vivo. This inhibitor had low nanomolar inhibitory potency (Ki=40 nM for Rgp, Ki=0.27 nM for Kgp) and good selectivity for host proteases and exhibited potent antibacterial activity against P. gingivalis by abrogating its manifold pathophysiological functions. The therapeutic potential of this inhibitor in vivo was also verified by suppressing the vascular permeability that was enhanced in guinea pigs by the organism and the gingival inflammation in beagle dog PD models. These findings suggest that a dual inhibitor of Rgp and Kgp would exhibit noteworthy anti-inflammatory activity in the treatment of PD.


Asunto(s)
Adhesinas Bacterianas/efectos de los fármacos , Cisteína Endopeptidasas/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/uso terapéutico , Oligopéptidos/uso terapéutico , Periodontitis/tratamiento farmacológico , Porphyromonas gingivalis/enzimología , Animales , Permeabilidad Capilar/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/toxicidad , Inhibidores de Cisteína Proteinasa/farmacología , Citocinas/metabolismo , Progresión de la Enfermedad , Perros , Evaluación Preclínica de Medicamentos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Cisteína-Endopeptidasas Gingipaínas , Cobayas , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Periodontitis/microbiología , Porphyromonas gingivalis/patogenicidad , Proteolisis , Especificidad por Sustrato , Virulencia
12.
Reumatismo ; 63(1): 29-37, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21509347

RESUMEN

OBJECTIVE: To assess the effect of caspase 3 inhibition, in the expression of intracellular antigens induced by apoptosis. MATERIAL AND METHODS: Skin explants of neonatal Balb/c mice were used to assess the autoantigen expression. Skin was obtained by punch biopsies, tissues were cultured in DMEM; cell death was induced by chemicals and assessed by TUNEL. The expression of La, Ro, Sm, RNP, Cajal Bodies and NuMa antigens were monitored by immunohistochemistry using autoantibodies or monoclonal antibodies against these antigens. RESULTS: Chemicals used to induce cell death, successfully produced apoptosis or necrosis in more than 60% of keratinocytes, and viability was significantly decreased when it was compared with those in controls. An increased expression of all skin intracellular antigens in skin biopsies treated with chemicals, major antigenic expression was detected with anti-La and anti-Ro antibodies. The caspase 3 inhibitor DEVD-CMK significantly decreased the expression of antigens induced by chemicals. CONCLUSION: By this result we can infer that caspase inhibitors modify apoptosis and decrease the autoantigens associated to cell death.


Asunto(s)
Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/inmunología , Autoantígenos/biosíntesis , Enfermedades Autoinmunes/prevención & control , Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/uso terapéutico , Piel/inmunología , Animales , Animales Recién Nacidos , Enfermedades Autoinmunes/etiología , Biopsia , Camptotecina/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/enzimología , Células Cultivadas/inmunología , Cicloheximida/farmacología , Inhibidores de Cisteína Proteinasa/farmacología , Evaluación Preclínica de Medicamentos , Peróxido de Hidrógeno/farmacología , Etiquetado Corte-Fin in Situ , Cloruro de Mercurio/farmacología , Ratones , Ratones Endogámicos BALB C , Técnicas de Cultivo de Órganos , Piel/enzimología
13.
Oncol Rep ; 25(3): 871-7, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21186403

RESUMEN

Propyl gallate (PG) used in processed food and medicinal preparations has been shown to induce cell death in normal and cancer cells. The inhibition of proteasome function has emerged as a useful strategy to maneuver apoptosis. Here, we investigated the combined effects of PG and MG132 (a proteasome inhibitor) on HeLa cells in relation to cell growth, cell death, reactive oxygen species (ROS) and glutathione (GSH). PG induced growth inhibition and apoptosis in HeLa cells, accompanied by the loss of mitochondrial membrane potential (MMP; ΔΨm), activation of caspase 3 and PARP cleavage. The levels of ROS and GSH depletion were increased in PG-treated HeLa cells. MG132 intensified apoptosis and PARP cleavage in PG-treated HeLa cells. MG132 also increased ROS levels including mitochondrial O2•-, MMP (ΔΨm) loss and GSH depletion in PG-treated HeLa cells. PG induced a G1 phase arrest of the cell cycle in HeLa cells, which was significantly prevented by MG132. MG132 alone inhibited HeLa cell growth via inducing the cell cycle arrests and triggering apoptosis. Conclusively, the inhibition of proteasome by MG132 plays a role as an enhancement factor in PG-induced apoptosis of HeLa cells via increasing ROS levels and GSH depletion.


Asunto(s)
Apoptosis/efectos de los fármacos , Leupeptinas/farmacología , Galato de Propilo/farmacología , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Ciclo Celular/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Inhibidores de Cisteína Proteinasa/uso terapéutico , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Glutatión/metabolismo , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Galato de Propilo/uso terapéutico , Inhibidores de Proteasoma , Especies Reactivas de Oxígeno/metabolismo
14.
Clin Exp Immunol ; 156(1): 172-82, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19220323

RESUMEN

Immunoproteasome up-regulation enhances the processing of nuclear factor-kappaB (NF-kappaB) and degradation of IkappaBalpha, which correlates with increased amounts of NF-kappaB in the various cells. Aberrant activation of NF-kappaB is involved in the pathogenesis of inflammatory bowel disease (IBD). The aim of this study was to elucidate the effect of proteasome inhibitor MG132 on experimental IBD. We investigated the effects of MG132 on intestinal inflammation and epithelial regeneration in both interleukin-10-deficient (IL-10(-/-)) mice and mice with dextran sulphate sodium (DSS)-induced colitis. Body weight, histological findings and tumour necrosis factor (TNF)-alpha mRNA expression, epithelial cell proliferation and NF-kappaB p65 activity in colonic tissues were examined. The effects of MG132 on cell proliferation, migration and multiple drug resistance 1 (MDR1) gene expression were determined in vitro. MG132 ameliorated intestinal inflammation of IL-10(-/-) mice by decreasing TNF-alpha mRNA expression in the colonic tissues, which was associated with suppression of NF-kappaB activation, and reduced significantly the number of Ki-67-positive intestinal epithelial cells. On the other hand, MG132 did not reduce intestinal inflammation in mice with DSS-induced colitis, and delayed significantly the recovery of body weight and epithelial regeneration. MG132 also suppressed significantly epithelial cell proliferation, cell migration and MDR1 gene expression in vitro. Proteasome inhibition reduces T cell-mediated intestinal inflammation, but may interrupt both epithelial regeneration and barrier function of colonic mucosa. Optimal use of proteasome inhibitor should be kept in mind when we consider its clinical application for patients with IBD.


Asunto(s)
Inhibidores de Cisteína Proteinasa/uso terapéutico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Leupeptinas/uso terapéutico , Animales , Proliferación Celular/efectos de los fármacos , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Interleucina-10/deficiencia , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
15.
Curr Opin Drug Discov Devel ; 11(5): 616-25, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18729013

RESUMEN

The proteasome is a multicatalytic protease complex that mediates the controlled degradation of intracellular proteins, including key components of pathways that contribute to cancer cell growth and immune cell signaling. Validation for the proteasome as a therapeutic target in oncology was provided by bortezomib, a proteasome inhibitor that was approved for the treatment of multiple myeloma in 2003. Since that time, a number of structurally and mechanistically distinct proteasome inhibitors have entered clinical development in oncology. In this review, the chemical properties, preclinical antitumor activities and early clinical trials of these next-generation proteasome inhibitors are described and the potential for future proteasome inhibitor development in autoimmune indications is discussed.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores de Cisteína Proteinasa/uso terapéutico , Diseño de Fármacos , Factores Inmunológicos/uso terapéutico , Inhibidores de Proteasoma , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/química , Enfermedades Autoinmunes/tratamiento farmacológico , Inhibidores de Cisteína Proteinasa/efectos adversos , Inhibidores de Cisteína Proteinasa/química , Evaluación Preclínica de Medicamentos , Humanos , Factores Inmunológicos/efectos adversos , Factores Inmunológicos/química , Estructura Molecular , Neoplasias/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Resultado del Tratamiento
16.
J Biol Chem ; 283(12): 7745-53, 2008 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-18184658

RESUMEN

Elucidation of Abeta-lowering agents that inhibit processing of the wild-type (WT) beta-secretase amyloid precursor protein (APP) site, present in most Alzheimer disease (AD) patients, is a logical approach for improving memory deficit in AD. The cysteine protease inhibitors CA074Me and E64d were selected by inhibition of beta-secretase activity in regulated secretory vesicles that produce beta-amyloid (Abeta). The regulated secretory vesicle activity, represented by cathepsin B, selectively cleaves the WT beta-secretase site but not the rare Swedish mutant beta-secretase site. In vivo treatment of London APP mice, expressing the WT beta-secretase site, with these inhibitors resulted in substantial improvement in memory deficit assessed by the Morris water maze test. After inhibitor treatment, the improved memory function was accompanied by reduced amyloid plaque load, decreased Abeta40 and Abeta42, and reduced C-terminal beta-secretase fragment derived from APP by beta-secretase. However, the inhibitors had no effects on any of these parameters in mice expressing the Swedish mutant beta-secretase site of APP. The notable efficacy of these inhibitors to improve memory and reduce Abeta in an AD animal model expressing the WT beta-secretase APP site present in the majority of AD patients provides support for CA074Me and E64d inhibitors as potential AD therapeutic agents.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/metabolismo , Catepsina B/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Leucina/análogos & derivados , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Catepsina B/genética , Bovinos , Inhibidores de Cisteína Proteinasa/uso terapéutico , Dipéptidos/uso terapéutico , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Expresión Génica , Humanos , Leucina/farmacología , Leucina/uso terapéutico , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Transgénicos
17.
Trends Pharmacol Sci ; 29(1): 22-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18037508

RESUMEN

Proteolytic activity is required for several key pro-tumorigenic processes: angiogenesis, invasion and metastasis. Consequently, increases in protease expression and activity are frequently reported in human cancers, and correlate with malignant progression and poor patient prognosis. Cysteine cathepsin proteases have recently emerged as an important class of proteolytic enzymes in cancer development, and cysteine cathepsin inhibitors have been proposed as anticancer agents. In this review, we highlight recent studies that now allow us to evaluate critically whether cysteine cathepsin inhibition represents a viable therapeutic strategy for the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Catepsinas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Inhibidores de Cisteína Proteinasa/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Neoplasias/fisiopatología , Regulación hacia Arriba/efectos de los fármacos
18.
Curr Med Chem ; 14(3): 289-314, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17305534

RESUMEN

Malaria is the most important parasitic disease worldwide, affecting more than 500 million people and causing close to 1 million deaths per annum. This serious fact is mainly attributable to the emergence of drug resistant strains of Plasmodium falciparum. The advances made in malaria chemotherapy based on unique aspects of the biochemistry and physiology of the responsible agents for this disease, parasites of Plasmodium genus, are covered in this review. Increasing resistance to conventional antimalarial drugs constitutes the main drawback for the persistence of this disease. In the present article, a comprehensive analysis of selected molecular targets is depicted in terms of their potential utility as chemotherapeutic agents. Our review focuses on different and important molecular targets for drug design that include proteases that hydrolyze hemoglobin, protein farnesyltransferase, heme detoxification pathway, polyamine pathways, dihydrofolate reductase, artemisinin-based combination therapies (ACTs), etc. Therefore, rational approaches to control malaria targeting metabolic pathways of malaria parasites which are essential for parasites survival are presented.


Asunto(s)
Antimaláricos/uso terapéutico , Malaria/tratamiento farmacológico , Transferasas Alquil y Aril/antagonistas & inhibidores , Animales , Artemisininas/uso terapéutico , Cisteína Endopeptidasas/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/uso terapéutico , Resistencia a Medicamentos , Ácidos Grasos/biosíntesis , Glutatión/metabolismo , Glutatión Transferasa/antagonistas & inhibidores , Hemoproteínas/efectos de los fármacos , Humanos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Poliaminas/metabolismo , Sesquiterpenos/uso terapéutico , Relación Estructura-Actividad , Tiorredoxinas/metabolismo
19.
Stroke ; 37(7): 1888-94, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16763180

RESUMEN

BACKGROUND AND PURPOSE: Matrix metalloproteinases (MMPs) and cysteine proteases (calpain and cathepsin B) play an important role in cell death and are upregulated after focal cerebral ischemia. Because there is a significant interaction between MMP-9 with calpain and cathepsin B, we investigated the role of E64d (a calpain and cathepsin B inhibitor) on MMP-9 activation in the rat focal ischemia model. METHODS: Male Sprague-Dawley rats were subjected to 2 hours of middle cerebral artery occlusion by using the suture insertion method followed by 22 hours of reperfusion. In the treatment group, a single dose of E64d (5 mg/kg IP) was administrated 30 minutes before the induction of focal ischemia, whereas the nontreatment group received dimethyl sulfoxide only. The neurological deficits, infarct volumes, Evans blue extravasation, brain edema, and MMP-9 activation in the brain were determined. RESULTS: Pretreatment with E64d produced a significant reduction in the cerebral infarction volume (353.1+/-19.8 versus 210.3+/-23.7 mm3) and the neurological deficits. Immunofluorescence studies showed MMP-9, calpain, and cathepsin B activation colocalized to both neurons and the neurovascular endothelial cells after ischemia, which was reduced by E64d. CONCLUSIONS: These results suggest that E64d treatment provides a neuroprotective effect to rats after transient focal cerebral ischemia by inhibiting the upregulation of MMP-9.


Asunto(s)
Calpaína/antagonistas & inhibidores , Catepsina B/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Leucina/análogos & derivados , Inhibidores de la Metaloproteinasa de la Matriz , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Fármacos Neuroprotectores/uso terapéutico , Premedicación , Animales , Barrera Hematoencefálica/efectos de los fármacos , Edema Encefálico/etiología , Edema Encefálico/patología , Hemorragia Cerebral/etiología , Inhibidores de Cisteína Proteinasa/administración & dosificación , Inhibidores de Cisteína Proteinasa/farmacología , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Azul de Evans/farmacocinética , Extravasación de Materiales Terapéuticos y Diagnósticos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/enzimología , Infarto de la Arteria Cerebral Media/patología , Leucina/administración & dosificación , Leucina/farmacología , Leucina/uso terapéutico , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Reperfusión , Método Simple Ciego
20.
Pancreas ; 29(2): 104-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15257101

RESUMEN

OBJECTIVES: Apoptosis appears in islets after isolation, and it has a detrimental effect on the islet function. To improve the outcome of clinical islet transplantation, it is crucial to protect islets from apoptosis. The aim of this study was to determine whether a caspase-3 inhibitor (Z-DEVD-FMK) added to culture media protects islets from apoptosis and to compare the effects of fetal bovine serum (FBS) with human serum albumin (HSA) as a protein supplement in culture. METHODS: Isolated human islets were cultured under 4 different conditions: 0.5% HSA (control), 0.5% HSA + 25 micromol/L Z-DEVD-FMK, 0.5% HSA + 100 micromol/L Z-DEVD-FMK and 10% FBS for 2 days. Next, 1000 IEQ islets precultured with 0.5% HSA and with or without 100 micromol/L Z-DEVD-FMK were transplanted to diabetic nude mice. RESULTS: The islet yields were higher in Z-DEVD-FMK-treated groups, and the inhibitor prevented apoptosis dose dependently. The yield and insulin release were higher in FBS-treated group than in the control group, but FBS did not affect apoptosis. All 6 mice transplanted with islets pretreated with Z-DEVD-FMK, and 3 of 8 mice with control islets became normoglycemic posttransplantation. CONCLUSION: Z-DEVD-FMK prevented apoptosis of isolated human islets and improved its function. FBS (10%) improved the islet yield and insulin secretion more than 0.5% HSA.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Diabetes Mellitus Experimental/cirugía , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/efectos de los fármacos , Oligopéptidos/farmacología , Adulto , Animales , Glucemia/análisis , Bovinos , Separación Celular , Células Cultivadas/efectos de los fármacos , Medios de Cultivo/farmacología , Inhibidores de Cisteína Proteinasa/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Sangre Fetal , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Riñón , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Oligopéptidos/uso terapéutico , Albúmina Sérica , Estreptozocina , Trasplante Heterólogo , Trasplante Heterotópico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA