Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Mol Neurobiol ; 57(5): 2206-2219, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-31981074

RESUMEN

Prion diseases are fatal infectious neurodegenerative disorders in human and animals caused by misfolding of the cellular prion protein (PrPC) into the infectious isoform PrPSc. These diseases have the potential to transmit within or between species, and no cure is available to date. Targeting the unfolded protein response (UPR) as an anti-prion therapeutic approach has been widely reported for prion diseases. Here, we describe the anti-prion effect of the chemical compound Sephin1 which has been shown to protect in mouse models of protein misfolding diseases including amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) by selectively inhibiting the stress-induced regulatory subunit of protein phosphatase 1, thus prolonging eIF2α phosphorylation. We show here that Sephin1 dose and time dependently reduced PrPSc in different neuronal cell lines which were persistently infected with various prion strains. In addition, prion seeding activity was reduced in Sephin1-treated cells. Importantly, we found that Sephin1 significantly overcame the endoplasmic reticulum (ER) stress induced in treated cells, as measured by lower expression of stress-induced aberrant prion protein. In a mouse model of prion infection, intraperitoneal treatment with Sephin1 significantly prolonged survival of prion-infected mice. When combining Sephin1 with the neuroprotective drug metformin, the survival of prion-infected mice was also prolonged. These results suggest that Sephin1 could be a potential anti-prion drug selectively targeting one component of the UPR pathway.


Asunto(s)
Guanabenzo/análogos & derivados , Proteínas PrPC/metabolismo , Proteínas PrPSc/metabolismo , Priones/efectos de los fármacos , Scrapie/tratamiento farmacológico , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Factor 2 Eucariótico de Iniciación/metabolismo , Guanabenzo/administración & dosificación , Guanabenzo/farmacología , Guanabenzo/uso terapéutico , Metformina/administración & dosificación , Metformina/farmacología , Metformina/uso terapéutico , Ratones , Neuroblastoma/patología , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 1/antagonistas & inhibidores , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Scrapie/patología
2.
J Biomol Struct Dyn ; 38(18): 5464-5473, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31820681

RESUMEN

Protein phosphatase 1 (PP1) is a critical regulator of several processes, such as muscle contraction, neuronal signaling, glycogen synthesis, and cell proliferation. Dysregulation of PP1 has recently been found to be implicated in cardiac dysfunctions, which indicates that PP1 could be an attractive therapeutic target. However, discovery of PP1 inhibitors with satisfied safety and efficiency is still a challenge. Here, in order to discover potential PP1 inhibitors, compounds extracted from traditional Chinese medicine (TCM) were screened by a novel integrated virtual screening protocol including pharmacophore modeling and docking approaches. Combined with protein phosphatase inhibition assay, ZINC43060554 showed strongly inhibitory activity with IC50 values of 26.78 µM. Furthermore, molecular dynamics simulation and Molecular Mechanics/Generalized Born Surface Area binding free-energy analysis were performed to examine the stability of ligand binding modes. These novel scaffolds discovered in the present study can be used for rational design of PP1 inhibitors with high affinity.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Medicina Tradicional China , Simulación del Acoplamiento Molecular , Proteína Fosfatasa 1 , Relación Estructura-Actividad Cuantitativa , Bioensayo , Simulación de Dinámica Molecular , Proteína Fosfatasa 1/antagonistas & inhibidores , Serina , Treonina
3.
Biomed Pharmacother ; 103: 838-843, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29710499

RESUMEN

This study was designed to investigate the effects of astragalosides on cardiac diastolic function, and an emphasis was placed on the variation of the upstream molecular regulators of phospholamban. Chronic heart failure (CHF) rats were induced by ligaturing the left anterior coronary artery, and rats in the therapeutic groups were treated with either a 50 mg/kg dose of captopril, 10 mg/kg dose of astragalosides or 20 mg/kg dose of astragalosides. Four weeks after treatment, the ratio of the early and atrial peak filling velocities (E/A) and maximal slope diastolic pressure decrement (-dp/dt) both decreased in CHF rats (by 30.3% and 25.5%, respectively) and significantly increased in 20 mg/kg astragalosides and captopril-treated rats. The protein phosphatase-1 activity was lower in the 20 mg/kg astragalosides group than in the CHF group (0.22 vs 0.44, P < 0.01), and the inhibitor-1 levels in the astragalosides and captopril-treated groups were increased. Chronic heart failure increased expression of protein kinase C-α and calcium-sensing receptor, and these changes were attenuated by astragalosides therapy. Astragalosides restored the diastolic dysfunction of chronic heart failure rats, possibly by downregulation of calcium-sensing receptor and protein kinase C-α, which in turn augmented inhibitor-1 expression, reduced protein phosphatase-1 activity and increased phospholamban phosphorylation.


Asunto(s)
Presión Sanguínea/fisiología , Insuficiencia Cardíaca/tratamiento farmacológico , Proteína Quinasa C-alfa/fisiología , Proteína Fosfatasa 1/fisiología , Receptores Sensibles al Calcio/fisiología , Saponinas/farmacología , Triterpenos/farmacología , Animales , Animales Recién Nacidos , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Diástole , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Insuficiencia Cardíaca/fisiopatología , Masculino , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Fosfatasa 1/antagonistas & inhibidores , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores Sensibles al Calcio/antagonistas & inhibidores , Saponinas/uso terapéutico , Triterpenos/uso terapéutico
4.
Biochem Pharmacol ; 109: 14-26, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27002182

RESUMEN

Cantharidin is a natural toxin and an active constituent in a traditional Chinese medicine used to treat tumors. Cantharidin acts as a semi-selective inhibitor of PPP-family ser/thr protein phosphatases. Despite sharing a common catalytic mechanism and marked structural similarity with PP1C, PP2AC and PP5C, human PP4C was found to be insensitive to the inhibitory activity of cantharidin. To explore the molecular basis for this selectivity, we synthesized and tested novel C5/C6-derivatives designed from quantum-based modeling of the interactions revealed in the co-crystal structures of PP5C in complex with cantharidin. Structure-activity relationship studies and analysis of high-resolution (1.25Å) PP5C-inhibitor co-crystal structures reveal close contacts between the inhibitor bridgehead oxygen and both a catalytic metal ion and a non-catalytic phenylalanine residue, the latter of which is substituted by tryptophan in PP4C. Quantum chemistry calculations predicted that steric clashes with the bulkier tryptophan side chain in PP4C would force all cantharidin-based inhibitors into an unfavorable binding mode, disrupting the strong coordination of active site metal ions observed in the PP5C co-crystal structures, thereby rendering PP4C insensitive to the inhibitors. This prediction was confirmed by inhibition studies employing native human PP4C. Mutation of PP5C (F446W) and PP1C (F257W), to mimic the PP4C active site, resulted in markedly suppressed sensitivity to cantharidin. These observations provide insight into the structural basis for the natural selectivity of cantharidin and provide an avenue for PP4C deselection. The novel crystal structures also provide insight into interactions that provide increased selectivity of the C5/C6 modifications for PP5C versus other PPP-family phosphatases.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/química , Cantaridina/química , Inhibidores Enzimáticos/química , Proteínas Nucleares/química , Fosfoproteínas Fosfatasas/química , Proteína Fosfatasa 1/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Cinética , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Unión Proteica , Dominios Proteicos , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Relación Estructura-Actividad
5.
FEBS J ; 281(19): 4519-34, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25132288

RESUMEN

The regulation of Plasmodium falciparum protein phosphatase type 1 (PfPP1) activity remains to be deciphered. Data from homologous eukaryotic type 1 protein phosphatases (PP1) suggest that several protein regulators should be involved in this essential process. One such regulator, named PfI2 based on its primary sequence homology with eukaryotic inhibitor 2 (I2), was recently shown to be able to interact with PfPP1 and to inhibit its phosphatase activity, mainly through the canonical 'RVxF' binding motif. The details of the structural and functional characteristics of this interaction are investigated here. Using NMR spectroscopy, a second site of interaction is suggested to reside between residues D94 and T117 and contains the 'FxxR/KxR/K' binding motif present in other I2 proteins. This site seems to play in concert/synergy with the 'RVxF' motif to bind PP1, because only mutations in both motifs were able to abolish this interaction completely. However, regarding the structure/function relationship, mutation of either the 'RVxF' or 'FxxR/KxR/K' motif is more drastic, because each mutation prevents the capacity of PfI2 to trigger germinal vesicle breakdown in microinjected Xenopus oocytes. This indicates that the tight association of the PfI2 regulator to PP1, mediated by a two-site interaction, is necessary to exert its function. Based on these results, the use of a peptide derived from the 'FxxR/KxR/K' PfI2 motif was investigated for its potential effect on Plasmodium growth. This peptide, fused at its N-terminus to a penetrating sequence, was shown to accumulate specifically in infected erythrocytes and to have an antiplasmodial effect.


Asunto(s)
Antimaláricos/química , Plasmodium falciparum/enzimología , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antimaláricos/metabolismo , Antimaláricos/farmacología , Células Cultivadas , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Eritrocitos/parasitología , Humanos , Datos de Secuencia Molecular , Plasmodium falciparum/efectos de los fármacos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/fisiología , Proteínas Protozoarias/química , Proteínas Protozoarias/fisiología , Xenopus laevis
6.
FEBS J ; 280(2): 612-26, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22260360

RESUMEN

Protein phosphatase-1 (PP1) and protein phosphatase-2A (PP2A) are responsible for the dephosphorylation of the majority of phosphoserine/threonine residues in cells. In this study, we show that (-)-epigallocatechin-3-gallate (EGCG) and 1,2,3,4,6-penta-O-galloyl-ß-D-glucose (PGG), polyphenolic constituents of green tea and tannins, inhibit the activity of the PP1 recombinant δ-isoform of the PP1 catalytic subunit and the native PP1 catalytic subunit (PP1c) with IC(50) values of 0.47-1.35 µm and 0.26-0.4 µm, respectively. EGCG and PGG inhibit PP2Ac less potently, with IC(50) values of 15 and 6.6 µm, respectively. The structure-inhibitory potency relationships of catechin derivatives suggests that the galloyl group may play a major role in phosphatase inhibition. The interaction of EGCG and PGG with PP1c was characterized by NMR and surface plasmon resonance-based binding techniques. Competitive binding assays and molecular modeling suggest that EGCG docks at the hydrophobic groove close to the catalytic center of PP1c, partially overlapping with the binding surface of microcystin-LR or okadaic acid. This hydrophobic interaction is further stabilized by hydrogen bonding via hydroxyl/oxo groups of EGCG to PP1c residues. Comparative docking shows that EGCG binds to PP2Ac in a similar manner, but in a distinct pose. Long-term treatment (24 h) with these compounds and other catechins suppresses the viability of HeLa cells with a relative effectiveness reminiscent of their in vitro PP1c-inhibitory potencies. The above data imply that the phosphatase-inhibitory features of these polyphenols may be implicated in the wide spectrum of their physiological influence.


Asunto(s)
Catequina/análogos & derivados , Taninos Hidrolizables/química , Proteína Fosfatasa 1/química , Estructura Terciaria de Proteína , Catequina/química , Catequina/metabolismo , Catequina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Células HeLa , Humanos , Taninos Hidrolizables/metabolismo , Taninos Hidrolizables/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo , Resonancia por Plasmón de Superficie , Té/química
7.
Toxicol Appl Pharmacol ; 265(2): 263-71, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23064102

RESUMEN

Blue-green algae (Spirulina sp., Aphanizomenon flos-aquae) and Chlorella sp. are commercially distributed as organic algae dietary supplements. Cyanobacterial dietary products in particular have raised serious concerns, as they appeared to be contaminated with toxins e.g. microcystins (MCs) and consumers repeatedly reported adverse health effects following consumption of these products. The aim of this study was to determine the toxin contamination and the in vitro cytotoxicity of algae dietary supplement products marketed in Germany. In thirteen products consisting of Aph. flos-aquae, Spirulina and Chlorella or mixtures thereof, MCs, nodularins, saxitoxins, anatoxin-a and cylindrospermopsin were analyzed. Five products tested in an earlier market study were re-analyzed for comparison. Product samples were extracted and analyzed for cytotoxicity in A549 cells as well as for toxin levels by (1) phosphatase inhibition assay (PPIA), (2) Adda-ELISA and (3) LC-MS/MS. In addition, all samples were analyzed by PCR for the presence of the mcyE gene, a part of the microcystin and nodularin synthetase gene cluster. Only Aph. flos-aquae products were tested positive for MCs as well as the presence of mcyE. The contamination levels of the MC-positive samples were ≤ 1 µg MC-LR equivalents g(-1) dw. None of the other toxins were found in any of the products. However, extracts from all products were cytotoxic. In light of the findings, the distribution and commercial sale of Aph. flos-aquae products, whether pure or mixed formulations, for human consumption appear highly questionable.


Asunto(s)
Aphanizomenon/química , Toxinas Bacterianas/análisis , Toxinas Bacterianas/toxicidad , Supervivencia Celular/efectos de los fármacos , Suplementos Dietéticos/análisis , Suplementos Dietéticos/toxicidad , Aphanizomenon/genética , Línea Celular , Cromatografía Liquida , ADN Bacteriano/química , ADN Bacteriano/genética , Ensayo de Inmunoadsorción Enzimática , Alemania , Humanos , Reacción en Cadena de la Polimerasa , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Espectrometría de Masas en Tándem
8.
Science ; 335(6065): 235-8, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22246779

RESUMEN

Painful stimuli activate nociceptive C fibers and induce synaptic long-term potentiation (LTP) at their spinal terminals. LTP at C-fiber synapses represents a cellular model for pain amplification (hyperalgesia) and for a memory trace of pain. µ-Opioid receptor agonists exert a powerful but reversible depression at C-fiber synapses that renders the continuous application of low opioid doses the gold standard in pain therapy. We discovered that brief application of a high opioid dose reversed various forms of activity-dependent LTP at C-fiber synapses. Depotentiation involved Ca(2+)-dependent signaling and normalization of the phosphorylation state of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. This also reversed hyperalgesia in behaving animals. Opioids thus not only temporarily dampen pain but may also erase a spinal memory trace of pain.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Potenciación a Largo Plazo/efectos de los fármacos , Fibras Nerviosas Amielínicas/efectos de los fármacos , Dolor Nociceptivo/tratamiento farmacológico , Piperidinas/administración & dosificación , Nervio Ciático/efectos de los fármacos , Sinapsis/efectos de los fármacos , Animales , Señalización del Calcio , Potenciales Evocados , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Masculino , Naloxona/administración & dosificación , Fibras Nerviosas Amielínicas/fisiología , Dolor Nociceptivo/fisiopatología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Remifentanilo , Nervio Ciático/fisiología , Somatostatina/administración & dosificación , Somatostatina/análogos & derivados , Médula Espinal/fisiología , Sinapsis/fisiología
9.
Brain Res ; 1371: 171-9, 2011 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-21112318

RESUMEN

An understanding of the diabetes-related changes in brain proteome composition in diabetes and treatment with Cynodon dactylon may provide insights to understand the brain function associated diabetes and metabolic protein mechanism responsible for this disease and in order to discover novel preventive and therapeutic drugs. We have performed a comprehensive proteomic analysis for comparison of rat brain proteome by using advance 2-dimensional electrophoresis (2DE) combined with mass spectrometry (MALDI-TOF-MS). We report here a comparison of alloxan-induced type 1 diabetic rats and C. dactylon-treated diabetic rats at 2 weeks following diabetes onset. As a result, we identified two differentially expressed proteins from rat brain. The identified proteins were functionally classified into two groups: (i) metabolic signalling protein (PPP1R14D) and ii) vesicle transport signalling protein (RAB18). This study provides a preliminary reference map of normal rat brain that will form a basis for comparative studies on normal and pathological conditions of the brain connected with diabetes and may serve as a potential tool for clinical diagnosis, therapeutics and prognosis and may provide new insights into novel mechanisms and therapeutic targets for diabetes-associated neurological disorder.


Asunto(s)
Química Encefálica , Cynodon/química , Diabetes Mellitus Experimental/metabolismo , Hipoglucemiantes/uso terapéutico , Proteínas del Tejido Nervioso/análisis , Fitoterapia , Extractos Vegetales/uso terapéutico , Hojas de la Planta/química , Proteína Fosfatasa 1/biosíntesis , Proteoma , Proteínas de Unión al GTP rab/biosíntesis , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Electroforesis en Gel Bidimensional , Etanol , Hipoglucemiantes/aislamiento & purificación , Modelos Biológicos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Filogenia , Extractos Vegetales/aislamiento & purificación , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/genética , Ratas , Homología de Secuencia de Aminoácido , Solventes , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteínas de Unión al GTP rab/genética
10.
Oncogene ; 29(47): 6233-44, 2010 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-20729910

RESUMEN

Here we report that ALDH1L1 (FDH, a folate enzyme with tumor suppressor-like properties) inhibits cell motility. The underlying mechanism involves F-actin stabilization, re-distribution of cytoplasmic actin toward strong preponderance of filamentous actin and formation of actin stress fibers. A549 cells expressing FDH showed a much slower recovery of green fluorescent protein-actin fluorescence in a fluorescence recovery after photobleaching assay, as well as an increase in G-actin polymerization and a decrease in F-actin depolymerization rates in pyren-actin fluorescence assays indicating the inhibition of actin dynamics. These effects were associated with robust dephosphorylation of the actin depolymerizing factor cofilin by PP1 and PP2A serine/threonine protein phosphatases, but not the cofilin-specific phosphatases slingshot and chronophin. In fact, the PP1/PP2A inhibitor calyculin prevented cofilin dephosphorylation and restored motility. Inhibition of FDH-induced apoptosis by the Jun N-terminal kinase inhibitor SP600125 or the pan-caspase inhibitor zVAD-fmk did not restore motility or levels of phosphor-cofilin, indicating that the observed effects are independent of FDH function in apoptosis. Interestingly, cofilin small interfering RNA or expression of phosphorylation-deficient S3A cofilin mutant resulted in a decrease of G-actin and the actin stress fiber formation, the effects seen upon FDH expression. In contrast, the expression of S3D mutant, mimicking constitutive phosphorylation, prevented these effects further supporting the cofilin-dependent mechanism. Dephosphorylation of cofilin and inhibition of motility in response to FDH can also be prevented by the increased folate in media. Furthermore, folate depletion itself, in the absence of FDH, resulted in cofilin dephosphorylation and inhibition of motility in several cell lines. Our experiments showed that these effects were folate specific and not a general response to nutrient starvation. Overall, this study shows the presence of distinct intracellular signaling pathways regulating motility in response to folate status and points toward mechanisms involving folates in promoting a malignant phenotype.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Movimiento Celular , Cofilina 1/metabolismo , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 2/metabolismo , Actinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Cofilina 1/deficiencia , Cofilina 1/genética , Citosol/efectos de los fármacos , Citosol/metabolismo , Suplementos Dietéticos , Inhibidores Enzimáticos/farmacología , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Mutación , Invasividad Neoplásica , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 2/antagonistas & inhibidores , Fibras de Estrés/efectos de los fármacos , Fibras de Estrés/metabolismo
11.
Mar Drugs ; 8(5): 1550-66, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20559488

RESUMEN

We report on screening tests of 66 extracts obtained from 35 marine sponge species from the Caribbean Sea (Curaçao) and from eight species from the Great Barrier Reef (Lizard Island). Extracts were prepared in aqueous and organic solvents and were tested for hemolytic, hemagglutinating, antibacterial and anti-acetylcholinesterase (AChE) activities, as well as their ability to inhibit or activate cell protein phosphatase 1 (PP1). The most interesting activities were obtained from extracts of Ircinia felix, Pandaros acanthifolium, Topsentia ophiraphidites, Verongula rigida and Neofibularia nolitangere. Aqueous and organic extracts of I. felix and V. rigida showed strong antibacterial activity. Topsentia aqueous and some organic extracts were strongly hemolytic, as were all organic extracts from I. felix. The strongest hemolytic activity was observed in aqueous extracts from P. acanthifolium. Organic extracts of N. nolitangere and I. felix inhibited PP1. The aqueous extract from Myrmekioderma styx possessed the strongest hemagglutinating activity, whilst AChE inhibiting activity was found only in a few sponges and was generally weak, except in the methanolic extract of T. ophiraphidites.


Asunto(s)
Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , Hemolíticos/farmacología , Poríferos/química , Acetilcolinesterasa/metabolismo , Animales , Antibacterianos/química , Australia , Región del Caribe , Inhibidores de la Colinesterasa/química , Inhibidores de la Colinesterasa/farmacología , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Hemaglutininas/química , Hemaglutininas/farmacología , Hemolíticos/química , Poríferos/clasificación , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteína Fosfatasa 1/metabolismo , Extractos de Tejidos/química , Extractos de Tejidos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA