Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Biochem Biophys Res Commun ; 696: 149504, 2024 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-38219489

RESUMEN

Regulated intramembrane proteolysis (RIP) is a two-step processing mechanism for transmembrane proteins consisting of ectodomain shedding (shedding), which removes the extracellular domain through juxtamembrane processing and intramembrane proteolysis, which processes membrane-anchored shedding products within the transmembrane domain. RIP irreversibly converts one transmembrane protein into multiple soluble proteins that perform various physiological functions. The only requirement for the substrate of γ-secretase, the major enzyme responsible for intramembrane proteolysis of type I transmembrane proteins, is the absence of a large extracellular domain, and it is thought that γ-secretase can process any type I membrane protein as long as it is shed. In the present study, we showed that the shedding susceptible type I membrane protein VIP36 (36 kDa vesicular integral membrane protein) and its homolog, VIPL, have different γ-secretase susceptibilities in their transmembrane domains. Analysis of the substitution mutants suggested that γ-secretase susceptibility is regulated by C-terminal amino acids in the transmembrane domain. We also compared the transmembrane domains of several shedding susceptible membrane proteins and found that each had a different γ-secretase susceptibility. These results suggest that the transmembrane domain is not simply a stretch of hydrophobic amino acids but is an important element that regulates membrane protein function by controlling the lifetime of the membrane-anchored shedding product.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide , Lectinas , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Lectinas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Dominios Proteicos , Membrana Celular/metabolismo
2.
Int J Mol Sci ; 25(12)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38928400

RESUMEN

The emergence of coronavirus disease 2019 (COVID-19) posed a major challenge to healthcare systems worldwide, especially as mutations in the culprit Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) complicated the development of vaccines and antiviral drugs. Therefore, the search for natural products with broad anti-SARS-CoV-2 capabilities is an important option for the prevention and treatment of similar infectious diseases. Lectins, which are widely recognized as antiviral agents, could contribute to the development of anti-SARS-CoV-2 drugs. This study evaluated the binding affinity of six lectins (including the cyanobacterial lectin from Microcystis viridis NIES-102 (MVL), and Jacalin, a lectin from the breadfruit, Artocarpus altilis) to the receptor binding domain (RBD) of the spike protein on the original (wild) SARS-CoV-2 and three of its mutants: Alpha, Delta, and Omicron. MVL and Jacalin showed distinct binding affinity to the RBDs of the four SARS-CoV-2 strains. The remaining four lectins (DB1, ConA, PHA-M and CSL3) showed no such binding affinity. Although the glycan specificities of MVL and Jacalin were different, they showed the same affinity for the spike protein RBDs of the four SARS-CoV-2 strains, in the order of effectiveness Alpha > Delta > original > Omicron. The verification of glycan-specific inhibition revealed that both lectins bind to RBDs by glycan-specific recognition, but, in addition, MVL binds to RBDs through protein-protein interactions.


Asunto(s)
Lectinas , Microcystis , Unión Proteica , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Lectinas/metabolismo , Lectinas/química , SARS-CoV-2/metabolismo , SARS-CoV-2/genética , Microcystis/metabolismo , Humanos , COVID-19/virología , COVID-19/metabolismo , Antivirales/farmacología , Antivirales/metabolismo , Dominios y Motivos de Interacción de Proteínas , Cianobacterias/metabolismo , Lectinas de Plantas/metabolismo , Lectinas de Plantas/química , Sitios de Unión , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Mutación
3.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36835396

RESUMEN

Amyloid-ß peptides (Aßs) are produced via cleavage of the transmembrane region of the amyloid precursor protein (APP) by γ-secretase and are responsible for Alzheimer's disease. Familial Alzheimer's disease (FAD) is associated with APP mutations that disrupt the cleavage reaction and increase the production of neurotoxic Aßs, i.e., Aß42 and Aß43. Study of the mutations that activate and restore the cleavage of FAD mutants is necessary to understand the mechanism of Aß production. In this study, using a yeast reconstruction system, we revealed that one of the APP FAD mutations, T714I, severely reduced the cleavage, and identified secondary APP mutations that restored the cleavage of APP T714I. Some mutants were able to modulate Aß production by changing the proportions of Aß species when introduced into mammalian cells. Secondary mutations include proline and aspartate residues; proline mutations are thought to act through helical structural destabilization, while aspartate mutations are thought to promote interactions in the substrate binding pocket. Our results elucidate the APP cleavage mechanism and could facilitate drug discovery.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Precursor de Proteína beta-Amiloide , Animales , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ácido Aspártico/genética , Mutación , Prolina/genética
4.
Int J Mol Sci ; 23(1)2022 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-35008932

RESUMEN

Amyloid beta peptides (Aßs) are generated from amyloid precursor protein (APP) through multiple cleavage steps mediated by γ-secretase, including endoproteolysis and carboxypeptidase-like trimming. The generation of neurotoxic Aß42/43 species is enhanced by familial Alzheimer's disease (FAD) mutations within the catalytic subunit of γ-secretase, presenilin 1 (PS1). FAD mutations of PS1 cause partial loss-of-function and decrease the cleavage activity. Activating mutations, which have the opposite effect of FAD mutations, are important for studying Aß production. Aph1 is a regulatory subunit of γ-secretase; it is presumed to function as a scaffold of the complex. In this study, we identified Aph1 mutations that are active in the absence of nicastrin (NCT) using a yeast γ-secretase assay. We analyzed these Aph1 mutations in the presence of NCT; we found that the L30F/T164A mutation is activating. When introduced in mouse embryonic fibroblasts, the mutation enhanced cleavage. The Aph1 mutants produced more short and long Aßs than did the wild-type Aph1, without an apparent modulatory function. The mutants did not change the amount of γ-secretase complex, suggesting that L30F/T164A enhances catalytic activity. Our results provide insights into the regulatory function of Aph1 in γ-secretase activity.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Endopeptidasas/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Mutación , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Dominio Catalítico , Endopeptidasas/metabolismo , Fibroblastos/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Presenilina-1/metabolismo , Proteolisis , Saccharomyces cerevisiae
5.
J Biol Chem ; 291(1): 435-46, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26559975

RESUMEN

γ-Secretase is a multisubunit membrane protein complex containing presenilin (PS1) as a catalytic subunit. Familial Alzheimer disease (FAD) mutations within PS1 were analyzed in yeast cells artificially expressing membrane-bound substrate, amyloid precursor protein, or Notch fused to Gal4 transcriptional activator. The FAD mutations, L166P and G384A (Leu-166 to Pro and Gly-384 to Ala substitution, respectively), were loss-of-function in yeast. We identified five amino acid substitutions that suppress the FAD mutations. The cleavage of amyloid precursor protein or Notch was recovered by the secondary mutations. We also found that secondary mutations alone activated the γ-secretase activity. FAD mutants with suppressor mutations, L432M or S438P within TMD9 together with a missense mutation in the second or sixth loops, regained γ-secretase activity when introduced into presenilin null mouse fibroblasts. Notably, the cells with suppressor mutants produced a decreased amount of Aß42, which is responsible for Alzheimer disease. These results indicate that the yeast system is useful to screen for mutations and chemicals that modulate γ-secretase activity.


Asunto(s)
Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Mutación/genética , Presenilina-1/genética , Supresión Genética , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Embrión de Mamíferos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ratones , Modelos Moleculares , Proteínas Mutantes/metabolismo , Presenilina-1/química , Estructura Terciaria de Proteína , Receptores Notch/metabolismo , Supresión Genética/efectos de los fármacos , beta-Galactosidasa/metabolismo
6.
EMBO J ; 31(4): 1014-27, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22157747

RESUMEN

Vesicle budding from the endoplasmic reticulum (ER) employs a cycle of GTP binding and hydrolysis to regulate assembly of the COPII coat. We have identified a novel mutation (sec24-m11) in the cargo-binding subunit, Sec24p, that specifically impacts the GTP-dependent generation of vesicles in vitro. Using a high-throughput approach, we defined genetic interactions between sec24-m11 and a variety of trafficking components of the early secretory pathway, including the candidate COPII regulators, Sed4p and Sec16p. We defined a fragment of Sec16p that markedly inhibits the Sec23p- and Sec31p-stimulated GTPase activity of Sar1p, and demonstrated that the Sec24p-m11 mutation diminished this inhibitory activity, likely by perturbing the interaction of Sec24p with Sec16p. The consequence of the heightened GTPase activity when Sec24p-m11 is present is the generation of smaller vesicles, leading to accumulation of ER membranes and more stable ER exit sites. We propose that association of Sec24p with Sec16p creates a novel regulatory complex that retards the GTPase activity of the COPII coat to prevent premature vesicle scission, pointing to a fundamental role for GTP hydrolysis in vesicle release rather than in coat assembly/disassembly.


Asunto(s)
Vesículas Cubiertas por Proteínas de Revestimiento/fisiología , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/fisiología , Proteínas de la Membrana/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Proteínas de la Membrana/química , Microscopía Electrónica , Microscopía Fluorescente , Modelos Moleculares , Proteínas de Saccharomyces cerevisiae/química , Técnicas del Sistema de Dos Híbridos
7.
Biochem Biophys Res Commun ; 478(4): 1751-7, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27608597

RESUMEN

The γ-secretase complex comprises presenilin (PS), nicastrin (NCT), anterior pharynx-defective 1 (Aph1), and presenilin enhancer 2 (Pen2). PS has two homologues, PS1 and PS2. Aph1 has two isoforms, Aph1a and Aph1b, with the former existing as two splice variants Aph1aL and Aph1aS. Each complex consists of one subunit each, resulting in six different γ-secretases. To better understand the functional differences among the γ-secretases, we reconstituted them using a yeast system and compared Notch1-cleavage and amyloid precursor protein (APP)-cleavage activities. Intriguingly, PS2/Aph1b had a clear substrate specificity: APP-Gal4, but not Notch-Gal4, was cleaved. In HEK cell lines expressing defined γ-secretase subunits, we showed that PS1/Aph1b, PS2/Aph1aL, PS2/Aph1aS and PS2/Aph1b γ-secretase produced amyloid ß peptide (Aß) with a higher Aß42+Aß43-to-Aß40 (Aß42(43)/Aß40) ratio than the other γ-secretases. In addition, PS2/Aph1aS γ-secretase produced less Notch intracellular domain (NICD) than did the other 5 γ-secretases. Considering that the Aß42(43)/Aß40 ratio is relevant in the pathogenesis of Alzheimer's disease (AD), and that inhibition of Notch cleavage causes severe side effect, these results suggest that the PS2/Aph1aS γ-secretase complex is a potential therapeutic target in AD.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/metabolismo , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/genética , Western Blotting , Endopeptidasas , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Fragmentos de Péptidos/metabolismo , Péptido Hidrolasas/genética , Presenilina-1/genética , Presenilina-2/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato
8.
Toxins (Basel) ; 15(8)2023 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-37624257

RESUMEN

A disintegrin and metalloproteinase (ADAM) family proteins are a major class of membrane-anchored multidomain proteinases that are responsible for the shedding of cell surface protein ectodomains, including amyloid precursor protein (APP). Human ADAM 9, 10, and 17 proteolyze APPs and produce non-amyloid-genic p3 peptides, instead of neurotoxic amyloid-ß peptides (Aßs; Aß40 and Aß42), which form fibrils and accumulate in the brain of patients with Alzheimer's disease (AD). The ADAM family is closely related to snake venom metalloproteinases (SVMPs), which are derived from ancestral ADAMs but act as soluble proteinases. To test the therapeutic potential of SVMPs, we purified SVMPs from Protobothrops flavoviridis venom using metal ion affinity and pooled into a cocktail. Thus, 9 out of 11 SVMPs in the P. flavoviridis genome were identified in the cocktail. SVMPs inhibited Aß secretion when added to human cell culture medium without affecting APP proteolysis. SVMPs degraded synthetic Aß40 and Aß42 peptides at the same cleavage site (α-site of APP) as ADAM9, 10, and 17. SVMPs did not degrade Aß fibrils but interfered with their formation, assessed using thioflavin-T. Thus, SVMPs have therapeutic potential for AD as an Aß-degrading protease, and the finding adds to the discovery of bioactive peptides from venoms as novel therapeutics.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Humanos , Ponzoñas , Proteolisis , Encéfalo , Proteínas de la Membrana , Proteínas ADAM
9.
J Biol Chem ; 286(52): 44569-75, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22074918

RESUMEN

γ-Secretase is composed of at least four proteins, presenilin (PS), nicastrin (NCT), Aph1, and Pen2. PS is the catalytic subunit of the γ-secretase complex, having aspartic protease activity. PS has two homologs, namely, PS1 and PS2. To compare the activity of these complexes containing different PSs, we reconstituted them in yeast, which lacks γ-secretase homologs. Yeast cells were transformed with PS1 or PS2, NCT, Pen2, Aph1, and artificial substrate C55-Gal4p. After substrate cleavage, Gal4p translocates to the nucleus and activates transcription of the reporter genes ADE2, HIS3, and lacZ. γ-Secretase activity was measured based on yeast growth on selective media and ß-galactosidase activity. PS1 γ-secretase was ∼24-fold more active than PS2 γ-secretase in the ß-galactosidase assay. Using yeast microsomes containing γ-secretase and C55, we compared the concentration of Aß generated by PS1 or PS2 γ-secretase. PS1 γ-secretase produced ∼24-fold more Aß than PS2 γ-secretase. We found the optimal pH of Aß production by PS2 to be 7.0, as for PS1, and that the PS2 complex included immature NCT, unlike the PS1 complex, which included mature NCT. In this study, we compared the activity of PS1 or PS2 per one γ-secretase complex. Co-immunoprecipitation experiments using yeast microsomes showed that PS1 concentrations in the γ-secretase complex were ∼28 times higher than that of PS2. Our data suggest that the PS1 complex is only marginally less active than the PS2 complex in Aß production.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Núcleo Celular/metabolismo , Complejos Multienzimáticos/metabolismo , Presenilina-1/metabolismo , Presenilina-2/metabolismo , Transporte Activo de Núcleo Celular/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/genética , Núcleo Celular/genética , Endopeptidasas , Regulación Fúngica de la Expresión Génica/genética , Humanos , Concentración de Iones de Hidrógeno , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microsomas/metabolismo , Complejos Multienzimáticos/genética , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Presenilina-1/genética , Presenilina-2/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
10.
Biochem Biophys Res Commun ; 417(1): 462-7, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22172944

RESUMEN

Cell adhesion molecule 1 (CADM1) is a type I transmembrane glycoprotein expressed in various tissues. CADM1 is a cell adhesion molecule with many functions, including roles in tumor suppression, apoptosis, mast cell survival, synapse formation, and spermatogenesis. CADM1 undergoes membrane-proximal cleavage called shedding, but the sheddase and mechanisms of CADM1 proteolysis have not been reported. We determined the cleavage site involved in CADM1 shedding by LC/MS/MS and showed that CADM1 shedding occurred in the membrane fraction and was inhibited by tumor necrosis factor-α protease inhibitor-1 (TAPI-1). An siRNA experiment revealed that ADAM10 mediates endogenous CADM1 shedding. In addition, the membrane-bound fragment generated by shedding was further cleaved by γ-secretase and generated CADM1-intracellular domain (ICD) in a mechanism called regulated intramembrane proteolysis (RIP). These results clarify the detailed mechanism of membrane-proximal cleavage of CADM1, suggesting the possibility of RIP-mediated CADM1 signaling.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Moléculas de Adhesión Celular/metabolismo , Desintegrinas/metabolismo , Inmunoglobulinas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Células COS , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular/genética , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inmunoglobulinas/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Ésteres del Forbol/farmacología , Inhibidores de Proteasas/farmacología
11.
J Neurosci Res ; 90(4): 870-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22183801

RESUMEN

Alzheimer's disease (AD) is characterized by senile plaques caused by amyloid-ß peptide (Aß) accumulation. It has been reported that Aß generation and accumulation occur in membrane microdomains, called lipid rafts, which are enriched in cholesterol and glycosphingolipids. Moreover, the ablation of cholesterol metabolism has been implicated in AD. Neprilysin (NEP), a neutral endopeptidase, is one of the major Aß-degrading enzymes in the brain. Activation of NEP is a possible therapeutic target. However, it remains unknown whether the activity of NEP is regulated by its association with lipid rafts. Here we show that only the mature form of NEP, which has been glycosylated in the Golgi, exists in lipid rafts, where it is directly associated with phosphatidylserine. Moreover, the localization of NEP in lipid rafts is enhanced by its dimerization, as shown using the NEP E403C homodimerization mutant. However, the protease activities of the mature form of NEP, as assessed by in vitro peptide hydrolysis, did not differ between lipid rafts and nonlipid rafts. We conclude that cholesterol and other lipids regulate the localization of mature NEP to lipid rafts, where the substrate Aß accumulates but does not modulate the protease activity of NEP.


Asunto(s)
Microdominios de Membrana/enzimología , Neprilisina/metabolismo , Péptidos beta-Amiloides/metabolismo , Línea Celular Transformada , Colesterol/metabolismo , Dimerización , Endopeptidasas/metabolismo , Humanos , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Mutación/genética , Neprilisina/genética , Transfección , beta-Ciclodextrinas/farmacología
12.
J Biol Chem ; 285(3): 1634-42, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19926793

RESUMEN

The amyloid-beta (Abeta) peptide, widely known as the causative molecule of Alzheimer disease (AD), is generated by the sequential cleavage of amyloid precursor protein (APP) by the aspartyl proteases BACE1/beta-secretase and presenilin/gamma-secretase. Inhibition of BACE1, therefore, is a promising strategy for preventing the progression of AD. However, beta-secretase inhibitors (BSIs) exhibit unexpectedly low potency in cells expressing "Swedish mutant" APP (APPswe) and in the transgenic mouse Tg2576, an AD model overexpressing APPswe. The Swedish mutation dramatically accelerates beta-cleavage of APP and hence the generation of Abeta; this acceleration has been assumed to underlie the poor inhibitory activity of BSI against APPswe processing. Here, we studied the mechanism by which the Swedish mutation causes this BSI potency decrease. Surprisingly, decreased BSI potency was not observed in an in vitro assay using purified BACE1 and substrates, indicating that the accelerated beta-cleavage resulting from the Swedish mutation is not its underlying cause. By focusing on differences between the cell-based and in vitro assays, we have demonstrated here that the potency decrease is caused by the aberrant subcellular localization of APPswe processing and not by accelerated beta-cleavage or the accumulation of the C-terminal fragment of beta-cleaved APP. Because most patients with sporadic AD express wild type APP, our findings suggest that the wild type mouse is superior to the Tg2576 mouse as a model for determining the effective dose of BSI for AD patients. This work provides novel insights into the potency decrease of BSI and valuable suggestions for its development as a disease-modifying agent.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Mutación , Inhibidores de Proteasas/farmacología , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide/aislamiento & purificación , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/química , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/aislamiento & purificación , Ácido Aspártico Endopeptidasas/metabolismo , Bovinos , Línea Celular Tumoral , Membrana Celular/metabolismo , Sistema Libre de Células/metabolismo , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo
13.
J Neurochem ; 116(1): 76-81, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21054383

RESUMEN

Dopamine receptor D(2) (DRD2) has two splicing isoforms, a long form (D2L) and short form (D2S), which have distinct functions in the dopaminergic system. However, the regulatory mechanism of the alternative splicing of DRD2 is unknown. In this study, we examined which splicing factors regulate the expression of D2L and D2S by over-expressing several RNA-binding proteins in HEK293 cells. In a cellular splicing assay, the over-expression of polypyrimidine tract-binding protein 1 (PTBP1) reduced the expression of D2S, whereas the knockdown of PTBP1 increased the expression of D2S. We also identified the regions of DRD2 that are responsive to PTBP1 using heterologous minigenes and deletion mutants. Our results indicate that PTBP1 regulates the alternative splicing of DRD2. Considering that DRD2 inhibits cAMP-dependent protein kinase A, which modulates the intracellular localization of PTBP1, PTBP1 may contribute to the autoregulation of DRD2 by regulating the expression of its isoforms.


Asunto(s)
Empalme Alternativo/genética , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Proteína de Unión al Tracto de Polipirimidina/fisiología , Receptores de Dopamina D2/genética , Línea Celular Tumoral , Antagonistas de los Receptores de Dopamina D2 , Técnicas de Silenciamiento del Gen , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/deficiencia , Humanos , Proteína de Unión al Tracto de Polipirimidina/deficiencia , Receptores de Dopamina D2/biosíntesis , Regulación hacia Arriba/genética
14.
Biosci Biotechnol Biochem ; 75(2): 396-400, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21307566

RESUMEN

The main signs of Alzheimer's disease (AD) are cognitive impairment and senile plaques composed of amyloid beta (Aß) observed in patients' brains. Therefore, therapy for AD focuses on the removal of Aß. We developed an "edible vaccine" that employs intestinal immunity with little to no side effects. Rice was utilized as an edible vaccine. It expressed GFP-Aß42. Aß rice was administered orally to wild-type (WT) mice causing production of anti-Aß antibodies. Since Aß rice was mixed with the cholera toxin B subunit (CTB), antibody against the rice seed protein was also produced. Then, mice were caused to develop immune tolerance against the rice seed protein by oral administration of Aß rice mixed with CTB. The results indicated that only anti-Aß antibodies were produced.


Asunto(s)
Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/inmunología , Anticuerpos/inmunología , Oryza/genética , Vacunas Comestibles/inmunología , Administración Oral , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Animales , Especificidad de Anticuerpos/inmunología , Mapeo Epitopo , Femenino , Expresión Génica , Masculino , Ratones , Plantas Modificadas Genéticamente/genética , Células Th2/inmunología , Vacunas Comestibles/administración & dosificación , Vacunas Comestibles/química , Vacunas Comestibles/genética
16.
Front Pharmacol ; 12: 766406, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34803710

RESUMEN

Snakebites are one of the major causes of death and long-term disability in the developing countries due to the presence of various bioactive peptides and proteins in snake venom. In Japan, the venom of the habu snake (Protobothrops flavoviridis) causes severe permanent damage due to its myonecrotic toxins. Antivenom immunoglobulins are an effective therapy for snakebites, and antivenom was recently developed with effective suppressive activity against myonecrosis induced by snake venom. To compare the properties of an antivenom having anti-myonecrotic activity with those of conventional antivenom with no anti-myonecrotic activity, this study applied focused proteomics analysis of habu venom proteins using 2D gel electrophoresis. As a target protein for antivenom immunoglobulins with anti-myonecrotic activity, we identified a thrombin-like serine protease, TLSP2 (TLf2), which was an inactive proteolytic isoform due to the replacement of the active site, His43 with Arg. Additionally, we identified the unique properties and a novel synergistic function of pseudoenzyme TLf2 as a myonecrosis-enhancing factor. To our knowledge, this is the first report of a function of a catalytically inactive snake serine protease.

17.
Biochem Biophys Res Commun ; 396(4): 927-32, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20460109

RESUMEN

ADAM family proteins are type I transmembrane, zinc-dependent metalloproteases. This family has multiple conserved domains, including a signal peptide, a pro-domain, a metalloprotease domain, a disintegrin (DI) domain, a cysteine-rich (Cys) domain, an EGF-like domain, a transmembrane domain, and a cytoplasmic domain. The Cys and DI domains may play active roles in regulating proteolytic activity or substrate specificity. ADAM19 has an autolytic processing activity within its Cys domain, and the processing is necessary for its proteolytic activity. To identify a new physiological function of ADAM19, we screened for associating proteins by using the extracellular domain of ADAM19 in a yeast two-hybrid system. Cysteine-rich protein 2 (CRIP2) showed an association with ADAM19 through its DI and Cys domains. Sequence analysis revealed that CRIP2 is a secretable protein without a classical signal. CRIP2 secretion was increased by overexpression of ADAM19 and decreased by suppression of ADAM19 expression. Moreover, CRIP2 secretion increased in parallel with the autolytic processing of ADAM19 stimulated by lipopolysaccharide. These findings suggest that ADAM19 autolysis is activated by lipopolysaccharide and that ADAM19 promotes the secretion of CRIP2.


Asunto(s)
Proteínas ADAM/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Lipopolisacáridos/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Activación Enzimática , Humanos , Proteínas con Dominio LIM , Estructura Terciaria de Proteína , Técnicas del Sistema de Dos Híbridos
18.
Biochem Biophys Res Commun ; 399(4): 593-9, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20682291

RESUMEN

Alzheimer's disease (AD) is pathologically characterized by the presence of extracellular senile plaques and intracellular neurofibrillary tangles. Amyloid beta-peptide (Abeta) is the main component of senile plaques, and the pathological load of Abeta in the brain has been shown to be a marker of the severity of AD. Abeta is produced from the amyloid precursor protein by membrane proteases and is known to aggregate. Recently, immune-mediated cerebral clearance of Abeta has been studied extensively as potential therapeutic strategy. In previous studies that used a purified Abeta challenge in a mouse model of AD, symptomatic improvement was reported. However, a clinical Alzheimer's vaccine trial in the United States was stopped because of severe side effects. Immunization with the strong adjuvant used in these trials might have activated an inflammatory Th1 response. In this study, to establish a novel, safer, lower-cost therapy for AD, we tested an oral vaccination in a wild-type and a transgenic mouse model of AD administered via green pepper leaves expressing GFP-Abeta. Anti-Abeta antibodies were effectively induced after oral immunization. We examined the immunological effects in detail and identified no inflammatory reactions. Furthermore, we demonstrated a reduction of Abeta in the immunized AD-model mice. These results suggest this edible vehicle for Abeta vaccination has a potential clinical application in the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/prevención & control , Vacunas contra el Alzheimer/administración & dosificación , Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/inmunología , Administración Oral , Péptidos beta-Amiloides/biosíntesis , Animales , Anticuerpos/metabolismo , Encéfalo/metabolismo , Capsicum/genética , Capsicum/metabolismo , Inmunoglobulina G/biosíntesis , Ratones , Ratones Transgénicos , Hojas de la Planta/genética , Hojas de la Planta/metabolismo , Vacunación
19.
Genes Cells ; 14(6): 751-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19486166

RESUMEN

In recent years, several novel types of disorder caused by the expansion of triplet repeats in specific genes have been characterized; in the "polyalanine diseases", these expanded repeats result in proteins with aberrantly elongated polyalanine tracts. In this study, we fused expanded polyalanine tracts to yellow fluorescent protein to examine their physical interaction with mitochondria. Tracts containing more than 23 alanine repeats were found to physically associate with mitochondria, strongly suggesting that an interaction between polyalanine tracts and mitochondria is a contributing factor in the pathology of polyalanine diseases. Furthermore, in in vitro experiments, polyalanine tracts induced release of cytochrome c from mitochondria and caspase-3 activation, independently of the mitochondrial permeability transition pore. These results suggest that oligomerized polyalanine tracts might induce the rupture of the mitochondrial membrane, the subsequent release of cytochrome c, and apoptosis. This novel mechanism for polyalanine tract cytotoxicity might be common to the pathogenesis of all polyalanine diseases. Further investigation of this mechanism might aid the development of therapies for these diseases.


Asunto(s)
Apoptosis/fisiología , Citocromos c/metabolismo , Mitocondrias , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Péptidos/metabolismo , Péptidos/farmacología , Animales , Células COS/ultraestructura , Caspasa 3/metabolismo , Chlorocebus aethiops , Activación Enzimática , Humanos , Ratones , Mitocondrias/enzimología , Mitocondrias/metabolismo , Mitocondrias Hepáticas/enzimología , Mitocondrias Hepáticas/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial
20.
Cells ; 9(2)2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32028683

RESUMEN

Amyloid-ß (Aß) peptides play a crucial role in the pathogenesis of Alzheimer's disease (AD). Aß production, aggregation, and clearance are thought to be important therapeutic targets for AD. Curcumin has been known to have an anti-amyloidogenic effect on AD. In the present study, we performed screening analysis using a curcumin derivative library with the aim of finding derivatives effective in suppressing Aß production with improved bioavailability of curcumin using CHO cells that stably express human amyloid-ß precursor protein and using human neuroblastoma SH-SY5Y cells. We found that the curcumin derivative GT863/PE859, which has been shown to have an inhibitory effect on Aß and tau aggregation in vivo, was more effective than curcumin itself in reducing Aß secretion. We further found that GT863 inhibited neither ß- nor γ-secretase activity, but did suppress γ-secretase-mediated cleavage in a substrate-dependent manner. We further found that GT863 suppressed N-linked glycosylation, including that of the γ-secretase subunit nicastrin. We also found that mannosidase inhibitors that block the mannose trimming step of N-glycosylation suppressed Aß production in a similar fashion, as was observed as a result of treatment with GT863. Collectively, these results suggest that GT863 downregulates N-glycosylation, resulting in suppression of Aß production without affecting secretase activity.


Asunto(s)
Péptidos beta-Amiloides/biosíntesis , Curcumina/análogos & derivados , Curcumina/farmacología , Alcaloides/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Células CHO , Cricetulus , Curcumina/química , Glicosilación , Humanos , Manosidasas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Notch/metabolismo , Especificidad por Sustrato , Swainsonina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA