Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Cell ; 39(2): 184-95, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20670888

RESUMEN

X-linked inhibitor of apoptosis (XIAP) is a potent antagonist of caspase apoptotic activity. XIAP also functions as an E3 ubiquitin ligase, targeting caspases for degradation. However, molecular pathways controlling XIAP activities remain unclear. Here, we report that nitric oxide (NO) reacts with XIAP by S-nitrosylating its RING domain (forming SNO-XIAP), thereby inhibiting E3 ligase and antiapoptotic activity. NO-mediated neurotoxicity and caspase activation have been linked to several neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's diseases. We find significant SNO-XIAP formation in brains of patients with these diseases, implicating this reaction in the etiology of neuronal damage. Conversely, S-nitrosylation of caspases is known to inhibit apoptotic activity. Unexpectedly, we find that SNO-caspase transnitrosylates (transfers its NO group) to XIAP, forming SNO-XIAP, and thus promotes cell injury and death. These findings provide insights into the regulation of caspase activation in neurodegenerative disorders mediated, at least in part, by nitrosative stress.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Óxido Nítrico/metabolismo , Procesamiento Proteico-Postraduccional , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Caspasas/genética , Activación Enzimática/genética , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Óxido Nítrico/genética , Estructura Terciaria de Proteína , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/genética
2.
Nature ; 457(7232): 1019-22, 2009 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-19118384

RESUMEN

The death inducing signalling complex (DISC) formed by Fas receptor, FADD (Fas-associated death domain protein) and caspase 8 is a pivotal trigger of apoptosis. The Fas-FADD DISC represents a receptor platform, which once assembled initiates the induction of programmed cell death. A highly oligomeric network of homotypic protein interactions comprised of the death domains of Fas and FADD is at the centre of DISC formation. Thus, characterizing the mechanistic basis for the Fas-FADD interaction is crucial for understanding DISC signalling but has remained unclear largely because of a lack of structural data. We have successfully formed and isolated the human Fas-FADD death domain complex and report the 2.7 A crystal structure. The complex shows a tetrameric arrangement of four FADD death domains bound to four Fas death domains. We show that an opening of the Fas death domain exposes the FADD binding site and simultaneously generates a Fas-Fas bridge. The result is a regulatory Fas-FADD complex bridge governed by weak protein-protein interactions revealing a model where the complex itself functions as a mechanistic switch. This switch prevents accidental DISC assembly, yet allows for highly processive DISC formation and clustering upon a sufficient stimulus. In addition to depicting a previously unknown mode of death domain interactions, these results further uncover a mechanism for receptor signalling solely by oligomerization and clustering events.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/química , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Agregación de Receptores , Transducción de Señal , Receptor fas/química , Receptor fas/metabolismo , Cristalografía por Rayos X , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/química , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Humanos , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo
3.
Br J Pharmacol ; 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38922847

RESUMEN

BACKGROUND AND PURPOSE: Inhibitors of voltage-gated sodium channels (NaVs) are important anti-epileptic drugs, but the contribution of specific channel isoforms is unknown since available inhibitors are non-selective. We aimed to create novel, isoform selective inhibitors of Nav channels as a means of informing the development of improved antiseizure drugs. EXPERIMENTAL APPROACH: We created a series of compounds with diverse selectivity profiles enabling block of NaV1.6 alone or together with NaV1.2. These novel NaV inhibitors were evaluated for their ability to inhibit electrically evoked seizures in mice with a heterozygous gain-of-function mutation (N1768D/+) in Scn8a (encoding NaV1.6) and in wild-type mice. KEY RESULTS: Pharmacologic inhibition of NaV1.6 in Scn8aN1768D/+ mice prevented seizures evoked by a 6-Hz shock. Inhibitors were also effective in a direct current maximal electroshock seizure assay in wild-type mice. NaV1.6 inhibition correlated with efficacy in both models, even without inhibition of other CNS NaV isoforms. CONCLUSIONS AND IMPLICATIONS: Our data suggest NaV1.6 inhibition is a driver of efficacy for NaV inhibitor anti-seizure medicines. Sparing the NaV1.1 channels of inhibitory interneurons did not compromise efficacy. Selective NaV1.6 inhibitors may provide targeted therapies for human Scn8a developmental and epileptic encephalopathies and improved treatments for idiopathic epilepsies.

4.
Clin Pharmacol Drug Dev ; 12(4): 447-456, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36530055

RESUMEN

Valbenazine and deutetrabenazine are vesicular monoamine transporter 2 (VMAT2) inhibitors approved for tardive dyskinesia. The clinical activity of valbenazine is primarily attributed to its only dihydrotetrabenazine (HTBZ) metabolite, [+]-α-HTBZ. Deutetrabenazine is a deuterated form of tetrabenazine and is metabolized to four deuterated HTBZ metabolites: [+]-α-deuHTBZ, [+]-ß-deuHTBZ, [-]-α-deuHTBZ, and [-]-ß-deuHTBZ. An open-label, crossover study characterized the pharmacokinetic profiles of the individual deuHBTZ metabolites, which have not been previously reported. VMAT2 inhibition and off-target interactions of the deuHTBZ metabolites were evaluated using radioligand binding. The only valbenazine HTBZ metabolite, [+]-α-HTBZ, was a potent VMAT2 inhibitor, with negligible affinity for off-target dopamine, serotonin, and adrenergic receptors. Following deutetrabenazine administration, [-]-α-deuHTBZ represented 66% of circulating deuHTBZ metabolites and was a relatively weak VMAT2 inhibitor with appreciable affinity for dopamine (D2S , D3 ) and serotonin (5-HT1A , 5-HT2B , 5-HT7 ) receptors. [+]-ß-deuHTBZ was the most abundant deuHTBZ metabolite that potently inhibited VMAT2, but it represented only 29% of total circulating deuHTBZ metabolites. The mean half-life of [+]-α-HTBZ (22.2 hours) was ∼3× longer than that of [+]-ß-deuHTBZ (7.7 hours). These findings are similar to studies with tetrabenazine, in that deutetrabenazine is metabolized to four deuHTBZ stereoisomers, the most abundant of which has negligible interaction with VMAT2 in vitro and appreciable affinity for several off-target receptors. In contrast, valbenazine's single HTBZ metabolite is a potent VMAT2 inhibitor in vitro with no discernible off-target activity. Determination of the effects of intrinsic/extrinsic variables on deutetrabenazine's safety/efficacy profile should incorporate assessment of the effects on all deuHTBZ metabolites.


Asunto(s)
Serotonina , Tetrabenazina , Humanos , Estudios Cruzados , Dopamina , Proteínas de Transporte Vesicular de Monoaminas
5.
Cancer Cell ; 5(1): 25-35, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14749124

RESUMEN

Apoptosis resistance commonly occurs in cancers, preventing activation of Caspase family cell death proteases. XIAP is an endogenous inhibitor of Caspases overexpressed in many cancers. We developed an enzyme derepression assay, based on overcoming XIAP-mediated suppression of Caspase-3, and screened mixture-based combinatorial chemical libraries for compounds that reversed XIAP-mediated inhibition of Caspase-3, identifying a class of polyphenylureas with XIAP-inhibitory activity. These compounds, but not inactive structural analogs, stimulated increases in Caspase activity, directly induced apoptosis of many types of tumor cell lines in culture, and sensitized cancer cells to chemotherapeutic drugs. Active compounds also suppressed growth of established tumors in xenograft models in mice, while displaying little toxicity to normal tissues. These findings validate IAPs as targets for cancer drug discovery.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Inhibidores Enzimáticos/farmacología , Neoplasias Experimentales/tratamiento farmacológico , Proteínas/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/metabolismo , Caspasa 3 , Técnicas Químicas Combinatorias , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Modelos Animales , Proteínas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Trasplante Heterólogo/patología , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X
6.
BMC Cancer ; 10: 11, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20067634

RESUMEN

BACKGROUND: The X-linked Inhibitor of Apoptosis (XIAP) has attracted much attention as a cancer drug target. It is the only member of the IAP family that can directly inhibit caspase activity in vitro, and it can regulate apoptosis and other biological processes through its C-terminal E3 ubiquitin ligase RING domain. However, there is controversy regarding XIAP's role in regulating tumor cell proliferation and survival under normal growth conditions in vitro. METHODS: We utilized siRNA to systematically knock down XIAP in ten human tumor cell lines and then monitored both XIAP protein levels and cell viability over time. To examine the role of XIAP in the intrinsic versus extrinsic cell death pathways, we compared the viability of XIAP depleted cells treated either with a variety of mechanistically distinct, intrinsic pathway inducing agents, or the canonical inducer of the extrinsic pathway, TNF-related apoptosis-inducing ligand (TRAIL). RESULTS: XIAP knockdown had no effect on the viability of six cell lines, whereas the effect in the other four was modest and transient. XIAP knockdown only sensitized tumor cells to TRAIL and not the mitochondrial pathway inducing agents. CONCLUSIONS: These data indicate that XIAP has a more central role in regulating death receptor mediated apoptosis than it does the intrinsic pathway mediated cell death.


Asunto(s)
Apoptosis , Regulación Neoplásica de la Expresión Génica , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Antineoplásicos/farmacología , Muerte Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Mitocondrias/metabolismo , Modelos Biológicos , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/química
7.
Biochem J ; 424(3): 335-45, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19788411

RESUMEN

The caspase-3 zymogen has essentially zero activity until it is cleaved by initiator caspases during apoptosis. However, a mutation of V266E in the dimer interface activates the protease in the absence of chain cleavage. We show that low concentrations of the pseudo-activated procaspase-3 kill mammalian cells rapidly and, importantly, this protein is not cleaved nor is it inhibited efficiently by the endogenous regulator XIAP (X-linked inhibitor of apoptosis). The 1.63 A (1 A = 0.1 nm) structure of the variant demonstrates that the mutation is accommodated at the dimer interface to generate an enzyme with substantially the same activity and specificity as wild-type caspase-3. Structural modelling predicts that the interface mutation prevents the intersubunit linker from binding in the dimer interface, allowing the active sites to form in the procaspase in the absence of cleavage. The direct activation of procaspase-3 through a conformational switch rather than by chain cleavage may lead to novel therapeutic strategies for inducing cell death.


Asunto(s)
Apoptosis , Caspasa 3/metabolismo , Precursores Enzimáticos/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/genética , Western Blotting , Caspasa 3/química , Caspasa 3/genética , Línea Celular , Cristalografía por Rayos X , Activación Enzimática , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Humanos , Modelos Moleculares , Mutación , Conformación Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Transfección , Proteína Inhibidora de la Apoptosis Ligada a X/genética
8.
Biochim Biophys Acta ; 1783(3): 448-54, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17996738

RESUMEN

Determination of the substrate specificity of site-specific proteases helps define their physiological roles. We developed a yeast-based system for defining the minimal substrate specificity of site-specific proteases, within the context of a protein. Using this system, we characterized the P4-P1 substrate specificity of the nematode apoptotic caspase CED-3. Apart from an absolute requirement for aspartate at the P1 position, CED-3 is a relatively promiscuous caspase capable of cleaving substrates bearing many amino acids at P4-P2 sites.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Caspasas/genética , Caspasas/metabolismo , Levaduras/genética , Animales , Ácido Aspártico/metabolismo , Sitios de Unión , Proteínas de Caenorhabditis elegans/análisis , Caspasas/análisis , Regulación de la Expresión Génica , Genes Reporteros , Modelos Biológicos , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de Proteína , Especificidad por Sustrato , Transcripción Genética , Transfección
9.
Methods Enzymol ; 446: 351-67, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18603133

RESUMEN

The measurement of general caspase activity and the quantification of purified recombinant caspases in vitro can be accomplished with relative ease. But the determination of which caspases are active in a cellular context is much more challenging. This is because commercially available small molecule substrates and inhibitors do not display sufficient specificity to dissect the complex interplay of caspase pathways. Here we describe procedures that can be used to validate which caspases are active in cell culture models and determine which caspases are responsible for specific cleavage events. We also recommend methods for working with recombinant initiator caspases in vitro and suggest ways to accurately assess the cleavage efficiency of natural caspase substrates.


Asunto(s)
Caspasas/metabolismo , Clorometilcetonas de Aminoácidos/metabolismo , Caspasa 8/metabolismo , Inhibidores de Caspasas , Caspasas/análisis , Células Cultivadas , Activación Enzimática , Humanos , Células Jurkat , Proteínas Recombinantes/análisis , Proteínas Recombinantes/metabolismo
10.
PLoS One ; 13(4): e0193236, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29608575

RESUMEN

Ozanimod (RPC1063) is a specific and potent small molecule modulator of the sphingosine 1-phosphate receptor 1 (S1PR1) and receptor 5 (S1PR5), which has shown therapeutic benefit in clinical trials of relapsing multiple sclerosis and ulcerative colitis. Ozanimod and its active metabolite, RP-101075, exhibit a similar specificity profile at the S1P receptor family in vitro and pharmacodynamic profile in vivo. The NZBWF1 mouse model was used in therapeutic dosing mode to assess the potential benefit of ozanimod and RP-101075 in an established animal model of systemic lupus erythematosus. Compared with vehicle-treated animals, ozanimod and RP-101075 reduced proteinuria over the duration of the study and serum blood urea nitrogen at termination. Additionally, ozanimod and RP-101075 reduced kidney disease in a dose-dependent manner, as measured by histological assessment of mesangial expansion, endo- and exo-capillary proliferation, interstitial infiltrates and fibrosis, glomerular deposits, and tubular atrophy. Further exploration into gene expression changes in the kidney demonstrate that RP-101075 also significantly reduced expression of fibrotic and immune-related genes in the kidneys. Of note, RP-101075 lowered the number of plasmacytoid dendritic cells, a major source of interferon alpha in lupus patients, and reduced all B and T cell subsets in the spleen. Given the efficacy demonstrated by ozanimod and its metabolite RP-101075 in the NZBWF1 preclinical animal model, ozanimod may warrant clinical evaluation as a potential treatment for systemic lupus erythematosus.


Asunto(s)
Indanos/farmacología , Inflamación/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Lupus Eritematoso Sistémico/patología , Oxadiazoles/farmacología , Receptores de Lisoesfingolípidos/metabolismo , Animales , ADN/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inmunoglobulina G/metabolismo , Inflamación/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/patología , Pruebas de Función Renal , Lupus Eritematoso Sistémico/tratamiento farmacológico , Ratones , Receptor de Interferón alfa y beta/metabolismo , Receptores de Esfingosina-1-Fosfato , Bazo/efectos de los fármacos , Bazo/patología
11.
J Biochem ; 142(4): 435-42, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17761692

RESUMEN

SerpinB6 (Proteinase inhibitor 6/PI-6) is an intracellular serpin produced by leukocytes, platelets, endothelial cells, keratinocytes and other epithelial cells. It is a potent cathepsin G inhibitor thought to protect monocytes, neutrophils and bystander cells from ectopic cathepsin G during inflammation. Here we show that serpinB6 also inhibits the human serine protease kallikrein-8 (hK8) and that in human and mouse skin, serpinB6 and kallikrein-8 co-localize in differentiated keratinocytes. SerpinB6 inhibits hK8 with an association rate constant (kass) of 1.8 +/- 0.2 x 10(5) M(-1)s(-1) compared to 3.4 +/- 0.2 x 10(6) M(-1) s(-1) for the interaction between the mouse orthologue of serpinB6 (SPI3/serpinb6a) and mouse kallikrein-8 (mK8). Molecular modelling suggested that the lower efficiency of the serpinB6/hK8 interaction is partly due to the bulkier P2 methionine residue of serpinB6 compared to the smaller P2 valine in SPI3. Taken together, these results suggest that serpinB6 is a physiologically relevant inhibitor of hK8 in skin. We postulate that serpinB6 protects the intracellular compartment of keratinocytes from ectopic hK8.


Asunto(s)
Calicreínas/antagonistas & inhibidores , Queratinocitos/enzimología , Serpinas/fisiología , Animales , Células Cultivadas , Humanos , Líquido Intracelular/enzimología , Líquido Intracelular/metabolismo , Calicreínas/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Serpinas/química
12.
Biosci Rep ; 34(2)2014 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27919034

RESUMEN

Caspase-2 is an evolutionarily conserved but enigmatic protease whose biological role remains poorly understood. To date, research into the functions of caspase-2 has been hampered by an absence of reagents that can distinguish its activity from that of the downstream apoptotic caspase, caspase-3. Identification of protein substrates of caspase-2 that are efficiently cleaved within cells may also provide clues to the role of this protease. We used a yeast-based transcriptional reporter system to define the minimal substrate specificity of caspase-2. The resulting profile enabled the identification of candidate novel caspase-2 substrates. Caspase-2 cleaved one of these proteins, the cancer-associated transcription factor Runx1, although with relatively low efficiency. A fluorogenic peptide was derived from the sequence most efficiently cleaved in the context of the transcriptional reporter. This peptide, Ac-VDTTD-AFC, was efficiently cleaved by purified caspase-2 and auto-activating caspase-2 in mammalian cells, and exhibited better selectivity for caspase-2 relative to caspase-3 than reagents that are currently available. We suggest that this reagent, used in parallel with the traditional caspase-3 substrate Ac-DEVD-AFC, will enable researchers to monitor caspase-2 activity in cell lysates and may assist in the determination of stimuli that activate caspase-2 in vivo.


Asunto(s)
Caspasa 2/química , Caspasa 2/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Péptidos/química , Péptidos/metabolismo , Caspasa 2/genética , Caspasa 3/química , Caspasa 3/genética , Caspasa 3/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Cisteína Endopeptidasas/genética , Activación Enzimática , Células HEK293 , Humanos , Péptidos/genética
13.
Science ; 335(6070): 851-5, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22344443

RESUMEN

The lyso-phospholipid sphingosine 1-phosphate modulates lymphocyte trafficking, endothelial development and integrity, heart rate, and vascular tone and maturation by activating G protein-coupled sphingosine 1-phosphate receptors. Here, we present the crystal structure of the sphingosine 1-phosphate receptor 1 fused to T4-lysozyme (S1P(1)-T4L) in complex with an antagonist sphingolipid mimic. Extracellular access to the binding pocket is occluded by the amino terminus and extracellular loops of the receptor. Access is gained by ligands entering laterally between helices I and VII within the transmembrane region of the receptor. This structure, along with mutagenesis, agonist structure-activity relationship data, and modeling, provides a detailed view of the molecular recognition and requirement for hydrophobic volume that activates S1P(1), resulting in the modulation of immune and stromal cell responses.


Asunto(s)
Receptores de Lisoesfingolípidos/química , Anilidas/química , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Muramidasa/química , Mutagénesis , Organofosfonatos/química , Conformación Proteica , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética
15.
Dev Cell ; 15(4): 497-508, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18854135

RESUMEN

The past decade and a half has witnessed the discovery of a large, evolutionarily conserved family of cellular genes bearing homology to the prototype baculovirus Inhibitor of Apoptosis (IAP). The logical decision in the field to also refer to these cellular proteins as IAPs fails to do justice to this versatile group of factors that play a wide range of roles in eukaryotic development and homeostasis which include, but are not limited to, the regulation of programmed cell death. Here we describe the shared functional characteristics of several well-characterized IAPs whose defining motifs place them more in the category of multifunctional modular protein interaction domains.


Asunto(s)
Apoptosis/fisiología , Evolución Molecular , Proteínas Inhibidoras de la Apoptosis/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Caspasas/metabolismo , Proteínas Inhibidoras de la Apoptosis/química , Proteínas Inhibidoras de la Apoptosis/genética , Modelos Biológicos , Datos de Secuencia Molecular , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Homología de Secuencia de Aminoácido
16.
J Biol Chem ; 283(28): 19499-510, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18458084

RESUMEN

Caspase-8 is the initiator caspase of the extrinsic apoptosis pathway and also has a role in non-apoptotic physiologies. Identifying endogenous substrates for caspase-8 by using integrated bioinformatics and biological approaches is required to delineate the diverse roles of this caspase. We describe a number of novel putative caspase-8 substrates using the Prediction of Protease Specificity (PoPS) program, one of which is histone deacetylase 7 (HDAC7). HDAC7 is cleaved faster than any other caspase-8 substrate described to date. It is also cleaved in primary CD4+CD8+ thymocytes undergoing extrinsic apoptosis. By using naturally occurring caspase inhibitors that have evolved exquisite specificity at concentrations found within the cell, we could unequivocally assign the cleavage activity to caspase-8. Importantly, cleavage of HDAC7 alters its subcellular localization and abrogates its Nur77 repressor function. Thus we demonstrate a direct role for initiator caspase-mediated proteolysis in promoting gene transcription.


Asunto(s)
Caspasa 8/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Represoras/metabolismo , Programas Informáticos , Transcripción Genética/fisiología , Animales , Apoptosis/fisiología , Células COS , Caspasa 8/química , Caspasa 8/genética , Inhibidores de Caspasas , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/química , Histona Desacetilasas/genética , Humanos , Ratones , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/química , Proteínas Represoras/genética , Especificidad por Sustrato/fisiología
17.
EMBO Rep ; 7(10): 988-94, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17016456

RESUMEN

Several of the inhibitor of apoptosis protein (IAP) family members regulate apoptosis in response to various cellular assaults. Some members are also involved in cell signalling, mitosis and targeting proteins to the ubiquitin-proteasome degradation machinery. The most intensively studied family member, X-linked IAP (XIAP), is a potent inhibitor of caspase activity; hence, it is generally assumed that direct caspase inhibition is an important conserved function of most members of the family. Biochemical and structural studies have precisely mapped the elements of XIAP required for caspase inhibition. Intriguingly, these elements are not conserved among IAPs. Here, we review current knowledge of the caspase-inhibitory potential of the human IAPs and show that XIAP is probably the only bona fide caspase inhibitor, suggesting that the other family members never gained the ability to directly inhibit caspase activity.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/fisiología , Proteína Inhibidora de la Apoptosis Ligada a X/fisiología , Secuencia de Aminoácidos , Inhibidores de Caspasas , Genes Ligados a X/fisiología , Glutatión Transferasa/genética , Humanos , Proteínas Inhibidoras de la Apoptosis/química , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/genética , Homología de Secuencia de Aminoácido , Proteína Inhibidora de la Apoptosis Ligada a X/química
18.
Mol Cell ; 23(4): 523-33, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16916640

RESUMEN

Caspase-7 is an obligate dimer of catalytic domains, with generation of activity requiring limited proteolysis within a region that separates the large and small chains of each domain. Using hybrid dimers we distinguish the relative contribution of each domain to catalysis by the whole molecule. We demonstrate that the zymogen arises from direct dimerization and not domain swapping. In contrast to previous conclusions, we show that only one of the catalytic domains must be proteolyzed to enable activation. The processed domain of this singly cleaved zymogen has the same catalytic activity as a domain of fully active caspase-7. A transient intermediate of singly cleaved dimeric caspase-7 can be found in a cell-free model of apoptosis induction. However, we see no evidence for an analogous intermediate of the related executioner caspase-3. Our study demonstrates the efficiency by which the executioner caspases are activated in vivo.


Asunto(s)
Caspasas/química , Caspasas/metabolismo , Ingeniería de Proteínas , Secuencia de Aminoácidos , Apoptosis , Ácido Aspártico/metabolismo , Caspasa 7 , Dominio Catalítico , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Dimerización , Activación Enzimática , Humanos , Células Jurkat , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
19.
EMBO J ; 24(3): 645-55, 2005 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-15650747

RESUMEN

The X-linked inhibitor of apoptosis protein (XIAP) uses its second baculovirus IAP repeat domain (BIR2) to inhibit the apoptotic executioner caspase-3 and -7. Structural studies have demonstrated that it is not the BIR2 domain itself but a segment N-terminal to it that directly targets the activity of these caspases. These studies failed to demonstrate a role of the BIR2 domain in inhibition. We used site-directed mutagenesis of BIR2 and its linker to determine the mechanism of executioner caspase inhibition by XIAP. We show that the BIR2 domain contributes substantially to inhibition of executioner caspases. A surface groove on BIR2, which also binds to Smac/DIABLO, interacts with a neoepitope generated at the N-terminus of the caspase small subunit following activation. Therefore, BIR2 uses a two-site interaction mechanism to achieve high specificity and potency for inhibition. Moreover, for caspase-7, the precise location of the activating cleavage is critical for subsequent inhibition. Since apical caspases utilize this cleavage site differently, we predict that the origin of the death stimulus should dictate the efficiency of inhibition by XIAP.


Asunto(s)
Inhibidores de Caspasas , Proteínas/química , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Sitios de Unión/genética , Caspasa 3 , Caspasa 7 , Caspasas/química , Caspasas/genética , Línea Celular , Secuencia Conservada , Evolución Molecular , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Proteínas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Proteína Inhibidora de la Apoptosis Ligada a X
20.
Biochemistry ; 43(32): 10560-9, 2004 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-15301553

RESUMEN

Caspase-14 is a developmentally regulated and tissue restricted member of the caspase family present in mammals. It is mainly found in epidermal keratinocytes and has been hypothesized to be involved in a tissue-specific form of cell senescence, leading to the differentiation of keratinocytes that form the cornified cell layer. However, the substrate specificity, activation mechanism, and function of this caspase have yet to be revealed. We report that caspase-14, in contrast to other caspases, is not produced in active form following expression in Escherichia coli but can be activated by high concentrations of kosmotropic salts. Moreover, proteolytic cleavage is also required since the kosmotropic salts were only effective on the cleaved enzyme. We propose that caspase-14 requires proteolytic cleavage within the catalytic domain, followed by dimerization and ordering of mobile active site loops, to generate a competent enzyme. In the presence of kosmotropic salt, we were able to determine the substrate specificities of mouse and human caspase-14. Surprisingly, the substrate preferences for the human and mouse enzyme are dissimilar. The results obtained with human caspase-14 classify this enzyme as a cytokine activator, but the mouse enzyme shows preferences similar to apical apoptotic caspases.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Células Cultivadas/enzimología , Activación Enzimática/fisiología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Caspasa 14 , Dimerización , Escherichia coli/enzimología , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA