Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Adv Drug Deliv Rev ; 157: 2-36, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32479930

RESUMEN

In its 33 years, ADDR has published regularly on the po5tential of oral delivery of biologics especially peptides and proteins. In the intervening period, analysis of the preclinical and clinical trial failures of many purported platform technologies has led to reflection on the true status of the field and reigning in of expectations. Oral formulations of semaglutide, octreotide, and salmon calcitonin have completed Phase III trials, with oral semaglutide being approved by the FDA in 2019. The progress made with oral peptide formulations based on traditional permeation enhancers is against a background of low and variable oral bioavailability values of ~1%, leading to a current perception that only potent peptides with a viable cost of synthesis can be realistically considered. Desirable features of candidates should include a large therapeutic index, some stability in the GI tract, a long elimination half-life, and a relatively low clearance rate. Administration in nanoparticle formats have largely disappointed, with few prototypes reaching clinical trials: insufficient particle loading, lack of controlled release, low epithelial particle uptake, and lack of scalable synthesis being the main reasons for discontinuation. Disruptive technologies based on engineered devices promise improvements, but scale-up and toxicology aspects are issues to address. In parallel, medicinal chemists are synthesizing stable hydrophobic macrocyclic candidate peptides of lower molecular weight and with potential for greater oral bioavailability than linear peptides, but perhaps without the same requirement for elaborate drug delivery systems. In summary, while there have been advances in understanding the limitations of peptides for oral delivery, low membrane permeability, metabolism, and high clearance rates continue to hamper progress.


Asunto(s)
Química Farmacéutica/métodos , Sistemas de Liberación de Medicamentos , Péptidos/administración & dosificación , Administración Oral , Animales , Disponibilidad Biológica , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Péptidos/química , Péptidos/farmacocinética , Proteínas/administración & dosificación , Proteínas/química , Proteínas/farmacocinética
2.
Sci Immunol ; 4(41)2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31757835

RESUMEN

MR1-restricted mucosal-associated invariant T (MAIT) cells play a unique role in the immune system. These cells develop intrathymically through a three-stage process, but the events that regulate this are largely unknown. Here, using bulk and single-cell RNA sequencing-based transcriptomic analysis in mice and humans, we studied the changing transcriptional landscape that accompanies transition through each stage. Many transcripts were sharply modulated during MAIT cell development, including SLAM (signaling lymphocytic activation molecule) family members, chemokine receptors, and transcription factors. We also demonstrate that stage 3 "mature" MAIT cells comprise distinct subpopulations including newly arrived transitional stage 3 cells, interferon-γ-producing MAIT1 cells and interleukin-17-producing MAIT17 cells. Moreover, the validity and importance of several transcripts detected in this study are directly demonstrated using specific mutant mice. For example, MAIT cell intrathymic maturation was found to be halted in SLAM-associated protein (SAP)-deficient and CXCR6-deficient mouse models, providing clear evidence for their role in modulating MAIT cell development. These data underpin a model that maps the changing transcriptional landscape and identifies key factors that regulate the process of MAIT cell differentiation, with many parallels between mice and humans.


Asunto(s)
Células T Invariantes Asociadas a Mucosa/inmunología , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Transcripción Genética/genética , Adulto , Animales , Diferenciación Celular/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología
3.
Pharmacol Res ; 117: 328-342, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27993717

RESUMEN

Proteinase-activated receptor 2 (PAR2) is a G protein-coupled receptor involved in metabolism, inflammation, and cancers. It is activated by proteolysis, which exposes a nascent N-terminal sequence that becomes a tethered agonist. Short synthetic peptides corresponding to this sequence also activate PAR2, while small organic molecules show promising PAR2 antagonism. Developing PAR2 ligands into pharmaceuticals is hindered by a lack of knowledge of how synthetic ligands interact with and differentially modulate PAR2. Guided by PAR2 homology modeling and ligand docking based on bovine rhodopsin, followed by cross-checking with newer PAR2 models based on ORL-1 and PAR1, site-directed mutagenesis of PAR2 was used to investigate the pharmacology of three agonists (two synthetic agonists and trypsin-exposed tethered ligand) and one antagonist for modulation of PAR2 signaling. Effects of 28 PAR2 mutations were examined for PAR2-mediated calcium mobilization and key mutants were selected for measuring ligand binding. Nineteen of twenty-eight PAR2 mutations reduced the potency of at least one ligand by >10-fold. Key residues mapped predominantly to a cluster in the transmembrane (TM) domains of PAR2, differentially influence intracellular Ca2+ induced by synthetic agonists versus a native agonist, and highlight subtly different TM residues involved in receptor activation. This is the first evidence highlighting the importance of the PAR2 TM regions for receptor activation by synthetic PAR2 agonists and antagonists. The trypsin-cleaved N-terminus that activates PAR2 was unaffected by residues that affected synthetic peptides, challenging the widespread practice of substituting peptides for proteases to characterize PAR2 physiology.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Péptidos/farmacología , Receptor PAR-2/metabolismo , Animales , Células CHO , Calcio/metabolismo , Bovinos , Línea Celular , Cricetulus , Humanos , Ligandos , Mutagénesis Sitio-Dirigida/métodos , Mutación/efectos de los fármacos , Dominios Proteicos/fisiología , Tripsina/metabolismo
4.
Bone ; 95: 162-174, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27913271

RESUMEN

Histone deacetylases (HDACs)2 play important roles in the epigenetic regulation of gene expression in cells and are emerging therapeutic targets for treating a wide range of diseases. HDAC inhibitors (HDACi)3 that act on multiple HDAC enzymes have been used clinically to treat a number of solid and hematological malignancies. HDACi are also currently being studied for their efficacy in non-malignant diseases, including pathologic bone loss, but this has necessitated a better understanding of the roles of individual HDAC enzymes, particularly the eleven zinc-containing isozymes. Selective isozyme-specific inhibitors currently being developed against class I HDACs (1, 2, 3 and 8) and class II HDACs (4, 5, 6, 7, 9 and 10) will be valuable tools for elucidating the roles played by individual HDACs in different physiological and pathological settings. Isozyme-specific HDACi promise to have greater efficacy and reduced side effects, as required for treating chronic disease over extended periods of time. This article reviews the current understanding of roles for individual HDAC isozymes and effects of HDACi on bone cells, (osteoblasts, osteoclasts and osteocytes), in relation to bone remodelling in conditions characterised by pathological bone loss, including periodontitis, rheumatoid arthritis and myeloma bone disease.


Asunto(s)
Remodelación Ósea/fisiología , Histona Desacetilasas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo
5.
Mucosal Immunol ; 10(1): 58-68, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27143301

RESUMEN

Despite recent breakthroughs in identifying mucosal-associated invariant T (MAIT) cell antigens (Ags), the precise requirements for in vivo MAIT cell responses to infection remain unclear. Using major histocompatibility complex-related protein 1 (MR1) tetramers, the MAIT cell response was investigated in a model of bacterial lung infection employing riboflavin gene-competent and -deficient bacteria. MAIT cells were rapidly enriched in the lungs of C57BL/6 mice infected with Salmonella Typhimurium, comprising up to 50% of αß-T cells after 1 week. MAIT cell accumulation was MR1-dependent, required Ag derived from the microbial riboflavin synthesis pathway, and did not occur in response to synthetic Ag, unless accompanied by a Toll-like receptor agonist or by co-infection with riboflavin pathway-deficient S. Typhimurium. The MAIT cell response was associated with their long-term accumulation in the lungs, draining lymph nodes and spleen. Lung MAIT cells from infected mice displayed an activated/memory phenotype, and most expressed the transcription factor retinoic acid-related orphan receptor γt. T-bet expression increased following infection. The majority produced interleukin-17 while smaller subsets produced interferon-γ or tumor necrosis factor, detected directly ex vivo. Thus the activation and expansion of MAIT cells coupled with their pro-inflammatory cytokine production occurred in response to Ags derived from microbial riboflavin synthesis and was augmented by co-stimulatory signals.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Pulmón/inmunología , Antígenos de Histocompatibilidad Menor/metabolismo , Membrana Mucosa/inmunología , Células T Asesinas Naturales/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Linfocitos T/inmunología , Animales , Antígenos Bacterianos/inmunología , Células Cultivadas , Receptores Coestimuladores e Inhibidores de Linfocitos T/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Riboflavina/biosíntesis , Riboflavina/inmunología , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Linfocitos T/microbiología , Factor de Necrosis Tumoral alfa/metabolismo
6.
Sci Rep ; 6: 37539, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27897196

RESUMEN

Dengue Virus (DENV) is the most prevalent global arbovirus, yet despite an increasing burden to health care there are currently no therapeutics available to treat infection. A potential target for antiviral drugs is the two-component viral protease NS2B-NS3pro, which is essential for viral replication. Interactions between the two components have been investigated here by probing the effect on the rate of enzyme catalysis of key mutations in a mobile loop within NS2B that is located at the interface of the two components. Steady-state kinetic assays indicated that the mutations greatly affect catalytic turnover. However, single turnover and fluorescence experiments have revealed that the mutations predominantly affect product release rather than substrate binding. Fluorescence analysis also indicated that the addition of substrate triggers a near-irreversible change in the enzyme conformation that activates the catalytic centre. Based on this mechanistic insight, we propose that residues within the mobile loop of NS2B control product release and present a new target for design of potent Dengue NS2B-NS3 protease inhibitors.


Asunto(s)
Virus del Dengue/química , Oligopéptidos/química , Serina Endopeptidasas/química , Proteínas no Estructurales Virales/química , Sustitución de Aminoácidos , Sitios de Unión , Biocatálisis , Clonación Molecular , Cristalografía por Rayos X , Virus del Dengue/enzimología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Cinética , Modelos Moleculares , Mutación , Oligopéptidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato , Termodinámica , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
7.
Br J Pharmacol ; 173(18): 2752-65, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27423137

RESUMEN

BACKGROUND AND PURPOSE: Diverse proteases cleave protease-activated receptor-2 (PAR2) on primary sensory neurons and epithelial cells to evoke pain and inflammation. Trypsin and tryptase activate PAR2 by a canonical mechanism that entails cleavage within the extracellular N-terminus revealing a tethered ligand that activates the cleaved receptor. Cathepsin-S and elastase are biased agonists that cleave PAR2 at different sites to activate distinct signalling pathways. Although PAR2 is a therapeutic target for inflammatory and painful diseases, the divergent mechanisms of proteolytic activation complicate the development of therapeutically useful antagonists. EXPERIMENTAL APPROACH: We investigated whether the PAR2 antagonist GB88 inhibits protease-evoked activation of nociceptors and protease-stimulated oedema and hyperalgesia in rodents. KEY RESULTS: Intraplantar injection of trypsin, cathespsin-S or elastase stimulated mechanical and thermal hyperalgesia and oedema in mice. Oral GB88 or par2 deletion inhibited the algesic and proinflammatory actions of all three proteases, but did not affect basal responses. GB88 also prevented pronociceptive and proinflammatory effects of the PAR2-selective agonists 2-furoyl-LIGRLO-NH2 and AC264613. GB88 did not affect capsaicin-evoked hyperalgesia or inflammation. Trypsin, cathepsin-S and elastase increased [Ca(2+) ]i in rat nociceptors, which expressed PAR2. GB88 inhibited this activation of nociceptors by all three proteases, but did not affect capsaicin-evoked activation of nociceptors or inhibit the catalytic activity of the three proteases. CONCLUSIONS AND IMPLICATIONS: GB88 inhibits the capacity of canonical and biased protease agonists of PAR2 to cause nociception and inflammation.


Asunto(s)
Inflamación/metabolismo , Nociceptores/metabolismo , Oligopéptidos/farmacología , Receptor PAR-2/agonistas , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligopéptidos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptor PAR-2/deficiencia , Receptor PAR-2/metabolismo , Relación Estructura-Actividad
8.
Protein Expr Purif ; 119: 124-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26647367

RESUMEN

Dengue Virus (DENV) infection is responsible for the world's most significant insect-borne viral disease. Despite an increasing global impact, there are neither prophylactic nor therapeutic options available for the effective treatment of DENV infection. An attractive target for antiviral drugs is the virally encoded trypsin-like serine protease (NS3pro) and its associated cofactor (NS2B). The NS2B-NS3pro complex is responsible for cleaving the viral polyprotein into separate functional viral proteins, and is therefore essential for replication. Recombinant expression of an active NS2B-NS3 protease has primarily been based on constructs linking the C-terminus of the approximately 40 amino acid hydrophilic cofactor domain of NS2B to the N-terminus of NS3pro via a flexible glycine linker. The resulting complex can be expressed in high yield, is soluble and catalytically active and has been used for most in vitro screening, inhibitor, and X-ray crystallographic studies over the last 15 years. Despite extensive analysis, no inhibitor drug candidates have been identified yet. Moreover, the effect of the artificial linker introduced between the protease and its cofactor is unknown. Two alternate methods for bacterial expression of non-covalently linked, catalytically active, NS2B-NS3pro complex are described here along with a comparison of the kinetics of substrate proteolysis and binding affinities of substrate-based aldehyde inhibitors. Both expression methods produced high yields of soluble protein with improved substrate proteolysis kinetics and inhibitor binding compared to their glycine-linked equivalent. The non-covalent association between NS2B and NS3pro is predicted to be more relevant for examining inhibitors that target cofactor-protease interactions rather than the protease active site. Furthermore, these approaches offer alternative strategies for the high yield co-expression of other protein assemblies.


Asunto(s)
Virus del Dengue/enzimología , Serina Endopeptidasas/biosíntesis , Proteínas no Estructurales Virales/biosíntesis , Antivirales/química , Cromatografía de Afinidad , Escherichia coli , Expresión Génica , Concentración de Iones de Hidrógeno , Cinética , Inhibidores de Proteasas/química , Serina Endopeptidasas/química , Serina Endopeptidasas/aislamiento & purificación , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/aislamiento & purificación
9.
Br J Pharmacol ; 171(17): 4112-24, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24821440

RESUMEN

BACKGROUND AND PURPOSE: Proteinase activated receptor 2 (PAR2) is a GPCR associated with inflammation, metabolism and disease. Clues to understanding how to block PAR2 signalling associated with disease without inhibiting PAR2 activation in normal physiology could be provided by studies of biased signalling. EXPERIMENTAL APPROACH: PAR2 ligand GB88 was profiled for PAR2 agonist and antagonist properties by several functional assays associated with intracellular G-protein-coupled signalling in vitro in three cell types and with PAR2-induced rat paw oedema in vivo. KEY RESULTS: In HT29 cells, GB88 was a PAR2 antagonist in terms of Ca(2+) mobilization and PKC phosphorylation, but a PAR2 agonist in attenuating forskolin-induced cAMP accumulation, increasing ERK1/2 phosphorylation, RhoA activation, myosin phosphatase phosphorylation and actin filament rearrangement. In CHO-hPAR2 cells, GB88 inhibited Ca(2+) release, but activated G(i/o) and increased ERK1/2 phosphorylation. In human kidney tubule cells, GB88 inhibited cytokine secretion (IL6, IL8, GM-CSF, TNF-α) mediated by PAR2. A rat paw oedema induced by PAR2 agonists was also inhibited by orally administered GB88 and compared with effects of locally administered inhibitors of G-protein coupled pathways. CONCLUSIONS AND IMPLICATIONS: GB88 is a biased antagonist of PAR2 that selectively inhibits PAR2/G(q/11)/Ca(2+)/PKC signalling, leading to anti-inflammatory activity in vivo, while being an agonist in activating three other PAR2-activated pathways (cAMP, ERK, Rho) in human cells. These findings highlight opportunities to design drugs to block specific PAR2-linked signalling pathways in disease, without blocking beneficial PAR2 signalling in normal physiology, and to dissect PAR2-associated mechanisms of disease in vivo.


Asunto(s)
Oligopéptidos/farmacología , Receptor PAR-2/antagonistas & inhibidores , Animales , Células CHO , Células Cultivadas , Cricetulus , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Células HT29 , Humanos , Ligandos , Oligopéptidos/administración & dosificación , Ratas , Receptor PAR-2/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
10.
Br J Pharmacol ; 171(5): 1180-94, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24354792

RESUMEN

Although it has been known since the 1960s that trypsin and chymotrypsin can mimic hormone action in tissues, it took until the 1990s to discover that serine proteinases can regulate cells by cleaving and activating a unique four-member family of GPCRs known as proteinase-activated receptors (PARs). PAR activation involves the proteolytic exposure of its N-terminal receptor sequence that folds back to function as a 'tethered' receptor-activating ligand (TL). A key N-terminal arginine in each of PARs 1 to 4 has been singled out as a target for cleavage by thrombin (PARs 1, 3 and 4), trypsin (PARs 2 and 4) or other proteases to unmask the TL that activates signalling via Gq , Gi or G12 /13 . Similarly, synthetic receptor-activating peptides, corresponding to the exposed 'TL sequences' (e.g. SFLLRN-, for PAR1 or SLIGRL- for PAR2) can, like proteinase activation, also drive signalling via Gq , Gi and G12 /13 , without requiring receptor cleavage. Recent data show, however, that distinct proteinase-revealed 'non-canonical' PAR tethered-ligand sequences and PAR-activating agonist and antagonist peptide analogues can induce 'biased' PAR signalling, for example, via G12 /13 -MAPKinase instead of Gq -calcium. This overview summarizes implications of this 'biased' signalling by PAR agonists and antagonists for the recognized roles the PARs play in inflammatory settings.


Asunto(s)
Receptores Proteinasa-Activados/metabolismo , Animales , Humanos , Inflamación/tratamiento farmacológico , Receptores Proteinasa-Activados/antagonistas & inhibidores , Transducción de Señal
11.
Br J Pharmacol ; 165(5): 1413-23, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21806599

RESUMEN

BACKGROUND AND PURPOSE: Many cells express proteinase activated receptor 2 (PAR2) on their plasma membrane. PAR2 is activated by proteolytic enzymes, such as trypsin and tryptase that cleave the receptor N-terminus, inititating signalling to intracellular G proteins. Studies on PAR2 have relied heavily upon activating effects of proteases and peptide agonists that lack stability and bioavailability in vivo. EXPERIMENTAL APPROACH: A novel small molecule agonist GB110 and an antagonist GB88 were characterized in vitro against trypsin, peptide agonists, PAR2 antibody, PAR1 agonists and flow cytometry,in seven cell lines using intracellular Ca(2+) mobilization and examined in vivo against PAR2- and PAR1-induced rat paw oedema. KEY RESULTS: GB110 is a potent non-peptidic agonist activating PAR2-mediated Ca(2+) release in HT29 cells (EC(50) ∼200 nM) and six other human cell lines, inducing PAR2 internalization. GB88 is a unique PAR2 antagonist, inhibiting PAR2 activated Ca(2+) release (IC(50) ∼2 µM) induced by native (trypsin) or synthetic peptide and non-peptide agonists. GB88 was a competitive and surmountable antagonist of agonist 2f-LIGRLO-NH(2), a competitive but insurmountable antagonist of agonist GB110, and a non-competitive insurmountable antagonist of trypsin. GB88 was orally active and anti-inflammatory in vivo, inhibiting acute rat paw oedema elicited by agonist GB110 and proteolytic or peptide agonists of PAR2 but not by corresponding agonists of PAR1 or PAR4. CONCLUSIONS AND IMPLICATIONS: The novel PAR2 agonist and antagonist modulate intracellular Ca(2+) and rat paw oedema, providing novel molecular tools for examining PAR2-mediated diseases.


Asunto(s)
Dipéptidos/farmacología , Isoxazoles/farmacología , Oligopéptidos/farmacología , Piperidinas/farmacología , Receptor PAR-2/agonistas , Receptor PAR-2/antagonistas & inhibidores , Compuestos de Espiro/farmacología , Animales , Antiinflamatorios/farmacología , Calcio/metabolismo , Línea Celular , Edema/metabolismo , Células HT29 , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Oligopéptidos/metabolismo , Péptidos/metabolismo , Ratas , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Tripsina/metabolismo
12.
J Periodontal Res ; 46(6): 697-703, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21745207

RESUMEN

BACKGROUND AND OBJECTIVE: Bone loss caused by enhanced osteoclast activity is a significant feature of periodontitis. Histone deacetylase inhibitors (HDACi) can suppress osteoclast-mediated bone loss in vitro and in vivo. This study investigated whether HDACi can suppress bone loss in experimental periodontitis. MATERIAL AND METHODS: Experimental periodontitis was induced in mice by oral inoculation with Porphyromonas gingivalis bacteria. Mice were treated orally with olive oil alone, with olive oil and a novel compound - 1179.4b - which targets both Class I and Class II histone deacetylases (HDACs) or with olive oil and MS-275, which targets Class I HDACs. Micro-computed tomography scans of live mice, stereo imaging and histological analyses were used to detect changes in bone. RESULTS: In the absence of treatment there was a 13.2% increase in bone volume in controls compared with a 7.4% decrease in P. gingivalis-inoculated mice. 1179.4b significantly reduced bone loss, with a 3.4% increase in bone volume (p < 0.01). MS-275 did not have a significant effect on P. gingivalis-induced bone loss. Histological analysis revealed that 1179.4b reduced bone loss despite having no effect on inflammation. CONCLUSION: HDACi were found to effectively suppress bone loss in the mouse model of periodontitis. 1179.4b - the inhibitor of Class I and Class II HDACs - was more effective at suppressing bone loss than MS-275, which targets Class I HDACs only. These compounds may therefore have the potential to be used for the management of periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar/enzimología , Pérdida de Hueso Alveolar/prevención & control , Inhibidores de Histona Desacetilasas/uso terapéutico , Pérdida de Hueso Alveolar/diagnóstico por imagen , Aminoquinolinas/uso terapéutico , Animales , Benzamidas/uso terapéutico , Densidad Ósea , Femenino , Ácidos Hidroxámicos/uso terapéutico , Imagenología Tridimensional , Ratones , Ratones Endogámicos BALB C , Aceite de Oliva , Osteoclastos/patología , Periodontitis/enzimología , Aceites de Plantas/uso terapéutico , Porphyromonas gingivalis , Piridinas/uso terapéutico , Microtomografía por Rayos X
13.
J Cell Physiol ; 226(12): 3233-41, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21344383

RESUMEN

Histone deacetylase inhibitors (HDACi) suppress cancer cell growth, inflammation, and bone resorption. The aim of this study was to determine the effect of inhibitors of different HDAC classes on human osteoclast activity in vitro. Human osteoclasts generated from blood mononuclear cells stimulated with receptor activator of nuclear factor kappa B (RANK) ligand were treated with a novel compound targeting classes I and II HDACs (1179.4b), MS-275 (targets class I HDACs), 2664.12 (targets class II HDACs), or suberoylanilide hydroxamic acid (SAHA; targets classes I and II HDACs). Osteoclast differentiation was assessed by expression of tartrate resistant acid phosphatase and resorption of dentine. Expression of mRNA encoding for osteoclast genes including RANK, calcitonin receptor (CTR), c-Fos, tumur necrosis factor (TNF) receptor associated factor (TRAF)6, nuclear factor of activated T cells (NFATc1), interferon-ß, TNF-like weak inducer of apoptosis (TWEAK), and osteoclast-associated receptor (OSCAR) were assessed. Expression of HDACs 1-10 during osteoclast development was also assessed. 1179.4b significantly reduced osteoclast activity (IC(50) < 0.16 nM). MS-275 (IC(50) 54.4 nM) and 2664.12 (IC(50) > 100 nM) were markedly less effective. A combination of MS-275 and 2664.12 inhibited osteoclast activity similar to 1179.4b (IC(50) 0.35 nM). SAHA was shown to suppress osteoclast activity (IC(50) 12 nM). 1179.4b significantly (P < 0.05) reduced NFATc1, CTR, and OSCAR expression during the later stages of osteoclast development. Class I HDAC 8 and Class II HDAC5 were both elevated (P < 0.05) during osteoclast development. Results suggest that inhibition of both classes I and II HDACs may be required to suppress human osteoclastic bone resorption in vitro.


Asunto(s)
Resorción Ósea/prevención & control , Diferenciación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Osteoclastos/efectos de los fármacos , Fosfatasa Ácida/genética , Benzamidas/farmacología , Resorción Ósea/enzimología , Resorción Ósea/patología , Células Cultivadas , Citocina TWEAK , Dentina/metabolismo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/farmacología , Interferón beta/genética , Isoenzimas/genética , Factores de Transcripción NFATC/genética , Osteoclastos/enzimología , Osteoclastos/patología , Proteínas Proto-Oncogénicas c-fos/genética , Piridinas/farmacología , Ligando RANK/metabolismo , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptores de Calcitonina/genética , Receptores de Superficie Celular/genética , Factor 6 Asociado a Receptor de TNF/genética , Fosfatasa Ácida Tartratorresistente , Factores de Tiempo , Factores de Necrosis Tumoral/genética , Vorinostat
14.
J Periodontal Res ; 44(3): 317-22, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19462493

RESUMEN

BACKGROUND AND OBJECTIVE: Live-animal micro-computed tomography is a new and promising technique that can be used to quantify changes in bone volume for periodontal disease models. The major aim of this study was to develop the methodology of live-animal micro-computed tomography and to determine the effect of a novel secretory phospholipase A2 inhibitor on alveolar bone loss. MATERIAL AND METHODS: Periodontitis was induced in mice by oral infection with Porphyromonas gingivalis over a period of 13 wk, and live-animal micro-computed tomography scans were taken at different time-points to determine bone volume changes with disease progression. This enabled conclusions to be made as to when treatment was most likely to be effective. In addition, the model was used to investigate a novel drug, the secretory phospholipase A2 inhibitor, KHO64, and its potential ability to inhibit osteoclast bone resorption and treat periodontitis. RESULTS: The results from live-animal micro-computed tomography scans revealed greater, statistically significant, bone volume loss in diseased mice compared with normal mice (p < 0.05). This corresponded to a larger area from the cemento-enamel junction to the alveolar bone crest, as assessed by stereo imaging (p < 0.001). These techniques can therefore detect and quantify alveolar bone loss. Both methods revealed that KHO64 had no significant effect on the volume of bone resorption. CONCLUSION: Live-animal micro-computed tomography is a robust, reproducible technique that clearly demonstrates significant time-dependent changes in alveolar bone volume in a small-animal model of periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar/diagnóstico por imagen , Pérdida de Hueso Alveolar/prevención & control , Infecciones por Bacteroides/enzimología , Inhibidores Enzimáticos/farmacología , Ácidos Pentanoicos/farmacología , Periodontitis/enzimología , Inhibidores de Fosfolipasa A2 , Microtomografía por Rayos X/métodos , Pérdida de Hueso Alveolar/enzimología , Animales , Densidad Ósea , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Femenino , Imagenología Tridimensional/métodos , Ratones , Ratones Endogámicos BALB C , Osteoclastos/efectos de los fármacos , Ácidos Pentanoicos/uso terapéutico , Periodontitis/microbiología , Periodontitis/prevención & control , Porphyromonas gingivalis , Cuello del Diente/diagnóstico por imagen
15.
Curr Med Chem ; 15(27): 2771-84, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18991636

RESUMEN

West Nile Virus (WNV) has spread rapidly during the last decade across five continents causing disease and fatalities in humans and mammals. It highlights the serious threat to both our health and the economy posed by viruses crossing species, in this case from migratory birds via mosquitoes to mammals. There is no vaccine or antiviral drug for treating WNV infection. One attractive target for antiviral development is a viral trypsin-like serine protease, encoded by the N-terminal 184 amino acids of NS3, which is only active when tethered to its cofactor, NS2B. This protease, NS2B/NS3pro, cleaves the viral polyprotein to release structural and non-structural viral proteins that are essential in viral replication and assembly of new virus particles. Disruption of this protease activity is lethal for virus replication. The NS3 protein also has other enzymes within its sequence (helicase, nucleoside triphosphatase, RNA triphosphatase), all of which are tightly regulated through localisation within membranous compartments in the infected cell. This review describes the various roles of NS3, focussing on NS2B-NS3 protease and its function and regulation in WNV replication and infection. Current advances towards development of antiviral inhibitors of NS2B/NS3pro are examined along with obstacles to their development as an antiviral therapy.


Asunto(s)
Antivirales/farmacología , Inhibidores de Proteasas/farmacología , Proteínas no Estructurales Virales/antagonistas & inhibidores , Virus del Nilo Occidental/enzimología , Antivirales/química , Antivirales/uso terapéutico , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteasas/química , Inhibidores de Proteasas/uso terapéutico , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/química , ARN Helicasas/metabolismo , Serina Endopeptidasas/química , Serina Endopeptidasas/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos , Fiebre del Nilo Occidental/tratamiento farmacológico
16.
Antimicrob Agents Chemother ; 52(4): 1454-61, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18212103

RESUMEN

The malaria parasite Plasmodium falciparum has at least five putative histone deacetylase (HDAC) enzymes, which have been proposed as new antimalarial drug targets and may play roles in regulating gene transcription, like the better-known and more intensively studied human HDACs (hHDACs). Fourteen new compounds derived from l-cysteine or 2-aminosuberic acid were designed to inhibit P. falciparum HDAC-1 (PfHDAC-1) based on homology modeling with human class I and class II HDAC enzymes. The compounds displayed highly potent antiproliferative activity against drug-resistant (Dd2) or drug sensitive (3D7) strains of P. falciparum in vitro (50% inhibitory concentration of 13 to 334 nM). Unlike known hHDAC inhibitors, some of these new compounds were significantly more toxic to P. falciparum parasites than to mammalian cells. The compounds inhibited P. falciparum growth in erythrocytes at both the early and late stages of the parasite's life cycle and caused altered histone acetylation patterns (hyperacetylation), which is a marker of HDAC inhibition in mammalian cells. These results support PfHDAC enzymes as being promising targets for new antimalarial drugs.


Asunto(s)
Aminoácidos Dicarboxílicos/farmacología , Antimaláricos/farmacología , Cisteína/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Plasmodium falciparum/efectos de los fármacos , Aminoácidos Dicarboxílicos/química , Animales , Antimaláricos/química , Cisteína/análogos & derivados , Cisteína/química , Resistencia a Medicamentos , Eritrocitos/parasitología , Humanos , Modelos Moleculares , Pruebas de Sensibilidad Parasitaria , Plasmodium falciparum/química , Plasmodium falciparum/enzimología , Homología de Secuencia de Aminoácido
17.
Artículo en Inglés | MEDLINE | ID: mdl-17846922

RESUMEN

Amyloids are filamentous protein deposits ranging in size from nanometres to microns and composed of aggregated peptide beta-sheets formed from parallel or anti-parallel alignments of peptide beta-strands. Amyloid-forming proteins have attracted a great deal of recent attention because of their association with over 30 diseases, notably neurodegenerative conditions like Alzheimer's, Huntington's, Parkinson's, Creutzfeldt-Jacob and prion disorders, but also systemic diseases such as amyotrophic lateral sclerosis (Lou Gehrig's disease) and type II diabetes. These diseases are all thought to involve important conformational changes in proteins, sometimes termed misfolding, that usually produce beta-sheet structures with a strong tendency to aggregate into water-insoluble fibrous polymers. Reasons for such conformational changes in vivo are still unclear. Intermediate aggregated state(s), rather than precipitated insoluble polymeric aggregates, have recently been implicated in cellular toxicity and may be the source of aberrant pathology in amyloid diseases. Numerous in vitro studies of short and medium length peptides that form amyloids have provided some clues to amyloid formation, with an alpha-helix to beta-sheet folding transition sometimes implicated as an intermediary step leading to amyloid formation. More recently, quite a few non-pathological amyloidogenic proteins have also been identified and physiological properties have been ascribed, challenging previous implications that amyloids were always disease causing. This article summarises a great deal of current knowledge on the occurrence, structure, folding pathways, chemistry and biology associated with amyloidogenic peptides and proteins and highlights some key factors that have been found to influence amyloidogenesis.


Asunto(s)
Amiloide/química , Péptidos/química , Proteínas/química , Animales , Cisteína/química , Humanos , Concentración de Iones de Hidrógeno , Sustancias Macromoleculares , Modelos Moleculares , Conformación Molecular , Enfermedades Neurodegenerativas/metabolismo , Conformación Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Electricidad Estática
18.
Br J Pharmacol ; 152(4): 429-48, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17603557

RESUMEN

Complement fragment (C)5a is a 74 residue pro-inflammatory polypeptide produced during activation of the complement cascade of serum proteins in response to foreign surfaces such as microorganisms and tissue damaged by physical or chemical injury. C5a binds to at least two seven-transmembrane domain receptors, C5aR (C5R1, CD88) and C5L2 (gpr77), expressed ubiquitously on a wide variety of cells but particularly on the surface of immune cells like macrophages, neutrophils and T cells. C5aR is a classical G protein-coupled receptor that signals through G alpha i and G alpha 16, whereas C5L2 does not appear to couple to G proteins and has no known signalling activity. Although C5a was first described as an anaphylatoxin and later as a leukocyte chemoattractant, the widespread expression of C5aR suggested more general functionality. Our understanding of the physiology of C5a has improved significantly in recent years through exploitation of receptor knockout and knocking mice, C5 and C5a antibodies, soluble recombinant C5a and C5a analogues and newly developed receptor antagonists. C5a is now also implicated in non-immunological functions associated with developmental biology, CNS development and neurodegeneration, tissue regeneration, and haematopoiesis. Combined receptor mutagenesis, molecular modelling, structure-activity relationship studies and species dependence for ligand potency on C5aR have been helpful for identifying ligand binding sites on the receptor and for defining mechanisms of receptor activation and inactivation. This review will highlight major developments in C5a receptor research that support C5aR as an important therapeutic target. The intriguing possibilities raised by the existence of a non-signalling C5a receptor are also discussed.


Asunto(s)
Complemento C5a/farmacología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/fisiología , Secuencia de Aminoácidos , Animales , Complemento C5a/química , Complemento C5a/uso terapéutico , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Estructura Molecular , Unión Proteica , Receptor de Anafilatoxina C5a/genética
19.
Arch Biochem Biophys ; 457(2): 187-96, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17184724

RESUMEN

A recombinant dengue 2 virus NS2B-NS3 protease (NS means non-structural virus protein) was compared with human furin for the capacity to process short peptide substrates corresponding to seven native substrate cleavage sites in the dengue viral polyprotein. Using fluorescence resonance energy transfer peptides to measure kinetics, the processing of these substrates was found to be selective for the Dengue protease. Substrates containing two or three basic amino acids (Arg or Lys) in tandem were found to be the best, with Abz-AKRRSQ-EDDnp being the most efficiently cleaved. The hydrolysis of dipeptide substrates Bz-X-Arg-MCA where X is a non-natural basic amino acid were also kinetically examined, the best substrates containing aliphatic basic amino acids. Our results indicated that proteolytic processing by dengue NS3 protease, tethered to its activating NS2B co-factor, was strongly inhibited by Ca2+ and kosmotropic salts of the Hofmeister's series, and significantly influenced by substrate modifications between S4 and S6'. Incorporation of basic non-natural amino acids in short peptide substrates had significant but differential effects on Km and k(cat), suggesting that further dissection of their influences on substrate affinity might enable the development of effective dengue protease inhibitors.


Asunto(s)
Aminoácidos Básicos/química , Colorantes Fluorescentes/química , Serina Endopeptidasas/química , Proteínas no Estructurales Virales/química , Sitios de Unión , Activación Enzimática , Transferencia Resonante de Energía de Fluorescencia , Furina/química , Concentración de Iones de Hidrógeno , Hidrólisis , Modelos Moleculares , Oligopéptidos/química , Proteínas Recombinantes/química , Sales (Química)/química , Especificidad por Sustrato
20.
Med Chem ; 2(2): 123-32, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16787361

RESUMEN

Here we describe the rational design, computer-aided virtual ligand docking and synthesis of 19 nonpeptidic compounds designed to inhibit histone deacetylases and kill melanoma cells. Compounds were derived from cysteine, fused at the S-terminus to 4-butanoyl hydroxamate, and at the N-terminus to 4-(dimethylamino)benzoic acid. The latter was extended by coupling to amines to form a small library of prospective anti-cancer compounds. Four compounds were cytotoxic at sub-micromolar concentrations against cells of a particularly aggressive human melanoma (MM96L), and nine compounds showed selectivities of >or=5:1 for killing human melanoma instead of normal human fibroblast cells. The most active compounds were shown to cause hyperacetylation of histones due to inhibition of histone deacetylases. Further refinement of these compounds may produce an anti-tumor drug suitable for treating melanoma.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Cisteína/farmacología , Inhibidores Enzimáticos/farmacología , Antineoplásicos/síntesis química , Línea Celular Tumoral , Cisteína/análogos & derivados , Cisteína/síntesis química , Diseño de Fármacos , Fibroblastos/citología , Fibroblastos/metabolismo , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Humanos , Melanoma/patología , Modelos Químicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...