Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 970: 176491, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38503399

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease with the hallmark of aggregation of beta-amyloid (Aß) into extracellular fibrillar deposition. Accumulating evidence suggests that soluble toxic Aß oligomers exert diverse roles in neuronal cell death, oxidative stress, neuroinflammation, and the eventual pathogenesis of AD. Aß is derived from the sequential cleavage of amyloid-ß precursor protein (APP) by ß-secretase (BACE1) and γ-secretase. The current effect of single targeting is not ideal for the treatment of AD. Therefore, developing multipotent agents with multiple properties, including anti-Aß generation and anti-Aß aggregation, is attracting more attention for AD treatment. Previous studies indicated that Quercetin was able to attenuate the effects of several pathogenetic factors in AD. Here, we showed that naturally synthesized Quercetin-3-O-glc-1-3-rham-1-6-glucoside (YCC31) could inhibit Aß production by reducing ß-secretase activity. Further investigations indicated that YCC31 could suppress toxic Aß oligomer formation by directly binding to Aß. Moreover, YCC31 could attenuate Aß-mediated neuronal death, ROS and NO production, and pro-inflammatory cytokines release. Taken together, YCC31 targeting multiple pathogenetic factors deserves further investigation for drug development of AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Humanos , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Quercetina/farmacología , Quercetina/uso terapéutico , Citocinas , Ácido Aspártico Endopeptidasas/metabolismo , Ácido Aspártico Endopeptidasas/uso terapéutico , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Glucósidos/uso terapéutico
2.
Cell Mol Immunol ; 18(8): 1883-1895, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32963355

RESUMEN

Rheumatoid arthritis (RA) is exacerbated by TNF-alpha signaling. However, it remains unclear whether TNF-α-activated TNFR1 and TNFR2 are regulated by extracellular factors. Here, we showed that soluble glycosylated interleukin-17 receptor D (sIL-17RD), which was produced by proteolytic cleavage, enhanced TNF-α-induced RA. We revealed that IL-17RD shedding was induced by the proteolytic enzyme TACE and enhanced by TNF-α expression in macrophages. Intriguingly, sIL-17RD was elevated in the sera of arthritic mice and rats. Recombinant sIL-17RD significantly enhanced the TNF-α-induced proinflammatory response by promoting TNF-α-TNFR-sIL-17RD complex formation and receptor clustering, leading to the accelerated development of collagen-induced arthritis. Our observations revealed that ectodomain shedding of IL-17RD occurred in RA to boost the TNF-α-induced inflammatory response. Targeting sIL-17RD may provide a new strategy for the therapy of RA.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Receptores de Interleucina-17 , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Artritis Reumatoide/metabolismo , Análisis por Conglomerados , Ratones , Ratas , Receptores de Interleucina-17/sangre , Factor de Necrosis Tumoral alfa/metabolismo
3.
Infect Immun ; 88(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32540868

RESUMEN

C-type lectin receptors (CLRs) play key roles in antifungal defense. CLR-induced NF-κB is central to CLR functions in immunity, and thus, molecules that control the amplitude of CLR-induced NF-κB could profoundly influence host defense against fungal pathogens. However, little is known about the mechanisms that negatively regulate CLR-induced NF-κB, and molecules which act on the CLR family broadly and which directly regulate acute CLR-signaling cascades remain unidentified. Here, we identify the ubiquitin-editing enzyme A20 as a negative regulator of acute NF-κB activation downstream of multiple CLR pathways. Absence of A20 suppression results in exaggerated CLR responses in cells which are A20 deficient and also cells which are A20 haplosufficient, including multiple primary immune cells. Loss of a single allele of A20 results in enhanced defense against systemic Candida albicans infection and prolonged host survival. Thus, A20 restricts CLR-induced innate immune responses in vivo and is a suppressor of host defense against systemic fungal infection.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Interacciones Microbiota-Huesped/inmunología , Lectinas Tipo C/inmunología , Procesamiento Proteico-Postraduccional , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Candida albicans/patogenicidad , Candidiasis/genética , Candidiasis/microbiología , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Femenino , Feto , Interacciones Microbiota-Huesped/genética , Inmunidad Innata , Lectinas Tipo C/genética , Hígado/inmunología , Hígado/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Cultivo Primario de Células , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/deficiencia , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Ubiquitina/genética , Ubiquitina/inmunología , Ubiquitinación
4.
Sci Adv ; 5(5): eaav7999, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31149635

RESUMEN

The mitochondrial antiviral signaling protein (MAVS) orchestrates host antiviral innate immune response to RNA virus infection. However, how MAVS signaling is controlled to eradicate virus while preventing self-destructive inflammation remains obscure. Here, we show that protein geranylgeranylation, a posttranslational lipid modification of proteins, limits MAVS-mediated immune signaling by targeting Rho family small guanosine triphosphatase Rac1 into the mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) at the mitochondria-ER junction. Protein geranylgeranylation and subsequent palmitoylation promote Rac1 translocation into MAMs upon viral infection. MAM-localized Rac1 limits MAVS' interaction with E3 ligase Trim31 and hence inhibits MAVS ubiquitination, aggregation, and activation. Rac1 also facilitates the recruitment of caspase-8 and cFLIPL to the MAVS signalosome and the subsequent cleavage of Ripk1 that terminates MAVS signaling. Consistently, mice with myeloid deficiency of protein geranylgeranylation showed improved survival upon influenza A virus infection. Our work revealed a critical role of protein geranylgeranylation in regulating antiviral innate immune response.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inmunidad Innata/fisiología , Neuropéptidos/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Prenilación de Proteína/inmunología , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Animales , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Masculino , Ratones Noqueados , Neuropéptidos/genética , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/mortalidad , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína RCA2 de Unión a GTP
5.
Theranostics ; 9(5): 1369-1384, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30867837

RESUMEN

Nuclear factor-kappa B (NF-κB) activation is critical for innate immune responses. However, cellular-intrinsic regulation of NF-κB activity during inflammatory diseases remains incompletely understood. Ubiquitin-like protein 4A (UBL4A, GdX) is a small adaptor protein involved in protein folding, biogenesis and transcription. Yet, whether GdX has a role during innate immune response is largely unknown. Methods: To investigate the involvement of GdX in innate immunity, we challenged GdX-deficient mice with lipopolysaccharides (LPS). To investigate the underlying mechanism, we performed RNA sequencing, real-time PCR, ELISA, luciferase reporter assay, immunoprecipitation and immunoblot analyses, flow cytometry, and structure analyses. To investigate whether GdX functions in inflammatory bowel disease, we generated dendritic cell (DC), macrophage (Mφ), epithelial-cell specific GdX-deficient mice and induced colitis with dextran sulfate sodium. Results: GdX enhances DC and Mφ-mediated innate immune defenses by positively regulating NF-κB signaling. GdX-deficient mice were resistant to LPS-induced endotoxin shock and DSS-induced colitis. DC- or Mφ- specific GdX-deficient mice displayed alleviated mucosal inflammation. The production of pro-inflammatory cytokines by GdX-deficient DCs and Mφ was reduced. Mechanistically, we found that tyrosine-protein phosphatase non-receptor type 2 (PTPN2, TC45) and protein phosphatase 2A (PP2A) form a complex with RelA (p65) to mediate its dephosphorylation whereas GdX interrupts the TC45/PP2A/p65 complex formation and restrict p65 dephosphorylation by trapping TC45. Conclusion: Our study provides a mechanism by which NF-κB signaling is positively regulated by an adaptor protein GdX in DC or Mφ to maintain the innate immune response. Targeting GdX could be a strategy to reduce over-activated immune response in inflammatory diseases.


Asunto(s)
Colitis/patología , Células Dendríticas/inmunología , Inmunidad Innata , Macrófagos/inmunología , FN-kappa B/metabolismo , Transducción de Señal , Ubiquitinas/metabolismo , Animales , Colitis/inducido químicamente , Sulfato de Dextran/administración & dosificación , Modelos Animales de Enfermedad , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Ubiquitinas/deficiencia
6.
Curr Alzheimer Res ; 15(9): 856-868, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29623840

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a neurodegenerative disease featured by memory loss, neuroinflammation and oxidative stress. Overproduction or insufficient clearance of Aß leads to its pathological aggregation and deposition, which is considered the predominant neuropathological hallmark of AD. Therefore, reducing Aß levels and inhibiting Aß-induced neurotoxicity are feasible therapeutic strategies for AD treatment. Wolfberry has been traditionally used as a natural antioxidant and anti-aging product. However, whether wolfberry species has therapeutic potential on AD remains unknown. METHOD: The effects of fruitless wolfberry-sprout extract (FWE) on Aß fibrillation and fibril disaggregation was measured by thioflavin T fluorescence and transmission electron microscope imaging; Aß oligomer level was determined by dot-blot; Cell viability and apoptosis was assessed by MTT and TUNEL assay. The levels of Aß40/42, oxidative stress biomarkers and inflammatory cytokines were detected by corresponding kits. 8-month-old male APP/PS1 mice and their age-matched WT littermates were treated with FWE or vehicle by oral administration (gavage) once a day for 4 weeks. Then the cognitive performance was determined using object recognition test and Y-maze test. The Aß burden and gliosis was evaluated by immunostaining and immunoblotting, respectively. RESULTS: FWE significantly inhibited Aß fibrillation and disaggregated the formed Aß fibrils, lowered Aß oligomer level and Aß-induced neuro-cytotoxicity, and attenuated oxidative stress in vitro. Oral administration of FWE remarkably improved cognitive function, reduced Aß burden, decreased gliosis and inflammatory cytokines release, and ameliorated oxidative stress in the brains of APP/PS1 mice. CONCLUSION: These findings indicate that FWE is a promising natural agent for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/etiología , Lycium/química , Extractos Vegetales/uso terapéutico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Interleucina-6/metabolismo , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Mutación/genética , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Presenilina-1/genética , Reconocimiento en Psicología/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
ACS Chem Neurosci ; 7(4): 505-18, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27015590

RESUMEN

Accumulating evidence suggested that soluble oligomeric ß-amyloid protein (Aß) exerts diverse roles in neuronal cell death, neuroinflammation, oxidative stress, and the eventual dementia associated with Alzheimer's disease (AD). Developing an agent with multiple properties may be a reasonable strategy for the treatment of AD. In this study, we isolated a novel multifunctional compound named camellikaempferoside B (YCF-2) from Fuzhuan brick tea. YCF-2 consists of kaempferol backbone, p-coumaric acid (p-CA) group, and a novel structure of rhamnopyranosyl group at the C-4' position, possessing the properties of both kaempferol and p-CA. YCF-2 significantly inhibited Aß production by decreasing ß-secretase activity. Moreover, YCF-2 suppressed Aß42 fibrillation and facilitated nontoxic oligomer formation by binding to Aß42 oligomers and by blocking the conformational transition to ß-sheet. Furthermore, YCF-2 ameliorated Aß-induced neuronal cell death, ROS production, inflammatory factor release, and microglia activation by blocking the NF-κB signaling pathway in microglia. These findings indicated that YCF-2 with a novel lead structure has potential applications for drug development for AD treatment.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Glicósidos/química , Glicósidos/farmacología , Fragmentos de Péptidos/metabolismo , Quercetina/análogos & derivados , Precursor de Proteína beta-Amiloide/genética , Animales , Células CHO , Muerte Celular/efectos de los fármacos , Células Cultivadas , Simulación por Computador , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Mutación/genética , Neuroglía/efectos de los fármacos , Neuroglía/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/ultraestructura , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Quercetina/química , Quercetina/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
8.
Behav Brain Res ; 296: 109-117, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26358659

RESUMEN

The pathologies of Alzheimer's disease (AD) is associated with soluble beta-amyloid (Aß) oligomers, neuroinflammation and oxidative stress. Decreasing the levels of Aß oligomer, glial activation and oxidative stress are potential therapeutic approaches for AD treatment. We previously found alpha-tocopherol quinine (α-TQ) inhibited Aß aggregation and cytotoxicity, decreased the release of inflammatory cytokines and reactive oxygen species (ROS) in vitro. However, whether α-TQ ameliorates memory deficits and other neuropathologies in mice or patients with AD remains unknown. In this study, we reported that orally administered α-TQ ameliorated memory impairment in APPswe/PS1dE9 transgenic mice, decreased oxidative stress and the levels of Aß oligomer in the brains of mice, prevented the production of inducible nitric oxide synthase and inflammatory mediators, such as interleukin-6 and interleukin-1ß, and inhibited microglial activation by inhibiting NF-κB signaling pathway. These findings suggest that α-TQ has potential therapeutic value for AD treatment.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antioxidantes/farmacología , Encéfalo/efectos de los fármacos , Inflamación/tratamiento farmacológico , Trastornos de la Memoria/tratamiento farmacológico , FN-kappa B/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Vitamina E/análogos & derivados , Péptidos beta-Amiloides , Animales , Antioxidantes/administración & dosificación , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Interleucinas/metabolismo , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Vitamina E/administración & dosificación , Vitamina E/farmacología
9.
J Biol Chem ; 290(2): 861-71, 2015 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-25378394

RESUMEN

TNF receptor 2 (TNFR2) exerts diverse roles in the pathogenesis of inflammatory and autoimmune diseases. Here, we report that TNFR2 but not TNFR1 forms a heteromer with interleukin-17 receptor D (IL-17RD), also named Sef, to activate NF-κB signaling. TNFR2 associates with IL-17RD, leading to mutual receptor aggregation and TRAF2 recruitment, which further activate the downstream cascade of NF-κB signaling. Depletion of IL-17RD impaired TNFR2-mediated activation of NF-κB signaling. Importantly, IL-17RD was markedly increased in renal tubular epithelial cells in nephritis rats, and a strong interaction of TNFR2 and IL-17RD was observed in the renal epithelia. The IL-17RD·TNFR2 complex in activation of NF-κB may explain the role of TNFR2 in inflammatory diseases including nephritis.


Asunto(s)
Inflamación/metabolismo , FN-kappa B/metabolismo , Nefritis/metabolismo , Receptores de Interleucina-17/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Inflamación/etiología , Inflamación/patología , Túbulos Renales Distales/metabolismo , Túbulos Renales Distales/patología , FN-kappa B/genética , Nefritis/etiología , Nefritis/patología , Multimerización de Proteína , Ratas , Receptores de Interleucina-17/química , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/química , Transducción de Señal/genética , Activación Transcripcional/genética
10.
Cell Signal ; 25(10): 2039-46, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23770285

RESUMEN

Sef (similar expression to fgf genes, also named IL-17RD) was identified as a negative regulator of fibroblast growth factor signaling. Sef-S, an alternative splice isoform of Sef, inhibits FGF-induced NIH3T3 cell proliferation. Here we report that Sef-S physically interacts with TAK1, induces Lys63-linked TAK1 polyubiquitination on lysine 209 and TAK1-mediated JNK and p38 activation. Co-overexpression of TAK1 WT, K34R, K150R, K158R mutants with Sef-S induces Lys63-linked TAK1 polyubiquitination whereas TAK1 K63R and K209R mutants fail. Furthermore, co-overexpression of Sef-S and TAK1 induce 293T cells apoptosis. These results reveal Sef-S actives Lys63-linked TAK1 polyubiquitination on lysine 209, induces TAK1-mediated JNK and p38 activation and also results apoptosis in 293T cells.


Asunto(s)
Apoptosis/genética , Interleucina-17/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Transducción de Señal , Empalme Alternativo/genética , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Interleucina-17/genética , Quinasas Quinasa Quinasa PAM/genética , Ratones , FN-kappa B/metabolismo , Células 3T3 NIH , Fosforilación , Ubiquitinación/genética
11.
PLoS One ; 7(11): e48540, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133641

RESUMEN

Amyloid precursor protein cleaving enzyme 1 (BACE1), an aspartyl protease, initiates processing of the amyloid precursor protein (APP) into ß-amyloid (Aß); the peptide likely contributes to development of Alzheimer's disease (AD). BACE1 is an attractive therapeutic target for AD treatment, but it exhibits other physiological activities and has many other substrates besides APP. Thus, inhibition of BACE1 function may cause adverse side effects. Here, we present a peptide, S1, isolated from a peptide library that selectively inhibits BACE1 hydrolytic activity by binding to the ß-proteolytic site on APP and Aß N-terminal. The S1 peptide significantly reduced Aß levels in vitro and in vivo and inhibited Aß cytotoxicity in SH-SY5Y cells. When applied to APPswe/PS1dE9 double transgenic mice by intracerebroventricular injection, S1 significantly improved the spatial memory as determined by the Morris Water Maze, and also attenuated their Aß burden. These results indicate that the dual-functional peptide S1 may have therapeutic potential for AD by both reducing Aß generation and inhibiting Aß cytotoxicity.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/química , Memoria , Péptidos/química , Conducta Espacial , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Benzotiazoles , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión/métodos , Biblioteca de Péptidos , Unión Proteica , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN , Sales de Tetrazolio/farmacología , Tiazoles/química , Tiazoles/farmacología
12.
Neurotoxicology ; 33(3): 482-90, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22445961

RESUMEN

Alzheimer's disease (AD) is a complex, multi-factorial neurodegenerative disease. The aggregation of soluble ß-amyloid (Aß) into fibrillar deposits is a pathological hallmark of AD. The Aß aggregate-induced neurotoxicity, inflammatory reactions, oxidative stress, and nitric oxide (NO) generation are strongly linked to the etiology of AD. Here, we show that the common dietary flavonoid, rutin, can dose-dependently inhibit Aß42 fibrillization and attenuate Aß42-induced cytotoxicity in SH-SY5Y neuroblastoma cells. Moreover, rutin decreases the formation of reactive oxygen species (ROS), NO, glutathione disulfide (GSSG), and malondialdehyde (MDA), reduces inducible nitric oxide synthase (iNOS) activity, attenuates mitochondrial damage, increases the glutathione (GSH)/GSSG ratio, enhances the activities of super oxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx), and modulates the production of proinflammatory cytokines by decreasing TNF-α and IL-1ß generation in microglia. Taken together, the actions of rutin on multiple pathogenic factors deserves further investigation for the prevention and treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/metabolismo , Rutina/farmacología , Catalasa/metabolismo , Línea Celular Tumoral , Citoprotección , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Interleucina-1beta/metabolismo , Malondialdehído/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
13.
Biochim Biophys Acta ; 1814(12): 1703-12, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21979582

RESUMEN

Increasing evidence indicates that amyloid aggregates, including oligomers, protofibrils or fibrils, are pivotal toxins in the pathogenesis of many amyloidoses such as Alzheimer's disease (AD), Parkinson's disease, Huntington's disease, prion-related diseases, type 2 diabetes and hereditary renal amyloidosis. Various oligomers assembled from different amyloid proteins share common structures and epitopes. Here we present data indicating that two oligomer-specific single chain variable fragment (scFv) antibodies isolated from a naïve human scFv library could conformation-dependently recognize oligomers assembled from α-synuclein, amylin, insulin, Aß1-40, prion peptide 106-126 and lysozyme, and fibrils from lysozyme. Further investigation showed that both scFvs inhibited the fibrillization of α-synuclein, amylin, insulin, Aß1-40 and prion peptide 106-126, and disaggregated their preformed fibrils. However, they both promoted the aggregation of lysozyme. Nevertheless, the two scFv antibodies could attenuate the cytotoxicity of all amyloids tested. Moreover, the scFvs recognized the amyloid oligomers in all types of plaques, Lewy bodies and amylin deposits in the brain tissues of AD and PD patients and the pancreas of type 2 diabetes patients respectively, and showed that most amyloid fibril deposits were colocalized with oligomers in the tissues. Such conformation-dependent scFv antibodies may have potential application in the investigation of aggregate structures, the mechanisms of aggregation and cytotoxicity of various amyloids, and in the development of diagnostic and therapeutic reagents for many amyloidoses.


Asunto(s)
Amiloide/inmunología , Amiloide/metabolismo , Amiloidosis/metabolismo , Dominios y Motivos de Interacción de Proteínas/inmunología , Anticuerpos de Cadena Única/metabolismo , Amiloide/química , Amiloidosis/patología , Reacciones Antígeno-Anticuerpo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Humanos , Cuerpos de Lewy/metabolismo , Cuerpos de Lewy/patología , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína/inmunología , Anticuerpos de Cadena Única/inmunología , Distribución Tisular , Células Tumorales Cultivadas
14.
Neurochem Int ; 57(8): 914-22, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20933033

RESUMEN

Alzheimer's disease (AD) is a complex, multifactorial neurodegenerative disease. The aggregation of beta-amyloid (Aß) into extracellular fibrillar deposition is a pathological hallmark of AD. The Aß aggregate-induced neurotoxicity, inflammatory reactions and oxidative stress are linked strongly to the etiology of AD. The currently available hitting-one-target drugs are insufficient for the treatment of AD. Therefore, finding multipotent agents able to modulate multiple targets simultaneously is attracting more attention. Previous studies indicated that vitamin E or its constituent such as α-tocopherol (α-T) was able to attenuate the effects of several pathogenetic factors in AD. However, ineffective or detrimental results were obtained from a number of clinical trials of vitamin E. Here, we showed that naturally synthesized RRR-α-tocopherol quinone (α-TQ), a main derivative of α-T, could inhibit Aß42 fibril formation dose-dependently. Further investigations indicated that α-TQ could attenuate Aß42-induced neurotoxicity toward SH-SY5Y neuroblastoma cells, disaggregate preformed fibrils and interfere with natural intracellular Aß oligomer formation. Moreover, α-TQ could decrease the formation of reactive oxygen species (ROS) and NO, and modulate the production of cytokines by decreasing TNF-α and IL-1ß and increasing IL-4 formation in microglia. Taken together, α-TQ targeting multiple pathogenetic factors deserves further investigation for prevention and treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Amiloide/antagonistas & inhibidores , Antioxidantes/farmacología , Citocinas/antagonistas & inhibidores , Mediadores de Inflamación/antagonistas & inhibidores , Óxido Nítrico/antagonistas & inhibidores , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Vitamina E/análogos & derivados , Amiloide/biosíntesis , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Antioxidantes/uso terapéutico , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Citocinas/fisiología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Humanos , Mediadores de Inflamación/fisiología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico/fisiología , Especies Reactivas de Oxígeno/metabolismo , Vitamina E/farmacología , Vitamina E/uso terapéutico
15.
J Alzheimers Dis ; 22(1): 107-17, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20847437

RESUMEN

Amyloid-ß (Aß) plays a pivotal role in Alzheimer's disease (AD) pathogenesis and in toxic mechanisms such as oxidative stress, mitochondrial dysfunction, calcium turbulence, and apoptosis induction. Therefore, interfering with Aß aggregation has long been one of the most promising strategies for AD treatment. Ecdysterones (ECRs) are steroidal hormones in insects and terrestrial plants that have high structural diversity and multiple beneficial pharmacological activities. Here, we studied the effects of six ECRs on Aß aggregation and cytotoxicity. Two ECRs with an acetoxyl group at the 2 or 3 position and saturated chains as side groups showed apparent promotion of Aß42 fibrilization, resulting in less Aß42 oligomers in the samples. Another three with unsaturated side chains clearly inhibited Aß aggregation and disaggregated preformed fibrils, but increased the Aß42 oligomer levels. Nevertheless, our MTT results showed that all ECRs tested inhibited Aß42-induced cytotoxicity. This protective activity may be partly attributable to ECR-mediated amelioration of A&beta42-induced release of reactive oxygen species. Taken together, our findings suggest that ECRs, a series of natural compounds in many plants and insects, have therapeutic potential in AD and that the deduced structure-activity relationships may be beneficial in drug design for the treatment of AD and other amyloidoses.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/toxicidad , Ecdisterona/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/toxicidad , Multimerización de Proteína/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Línea Celular Tumoral , Ecdisterona/química , Ecdisterona/uso terapéutico , Humanos , Fragmentos de Péptidos/metabolismo , Pliegue de Proteína/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
16.
Biochem Biophys Res Commun ; 390(4): 1250-4, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19878655

RESUMEN

Smaller, soluble oligomers of beta-amyloid (Abeta) play a critical role in the pathogenesis of Alzheimer's disease (AD). Selective inhibition of Abeta oligomer formation provides an optimum target for AD therapy. Some polyphenols have potent anti-amyloidogenic activities and protect against Abeta neurotoxicity. Here, we tested the effects of ellagic acid (EA), a polyphenolic compound, on Abeta42 aggregation and neurotoxicity in vitro. EA promoted Abeta fibril formation and significant oligomer loss, contrary to previous results that polyphenols inhibited Abeta aggregation. The results of transmission electron microscopy (TEM) and Western blot displayed more fibrils in Abeta42 samples co-incubated with EA in earlier phases of aggregation. Consistent with the hypothesis that plaque formation may represent a protective mechanism in which the body sequesters toxic Abeta aggregates to render them harmless, our MTT results showed that EA could significantly reduce Abeta42-induced neurotoxicity toward SH-SY5Y cells. Taken together, our results suggest that EA, an active ingredient in many fruits and nuts, may have therapeutic potential in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Ácido Elágico/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Línea Celular , Humanos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Estructura Secundaria de Proteína/efectos de los fármacos
17.
Neurotoxicology ; 30(6): 986-95, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19744518

RESUMEN

Beta-amyloid (Abeta) aggregation has been strongly associated with the neurodegenerative pathology and a cascade of harmful event rated to Alzheimer's disease (AD). Inhibition of Abeta assembly, destabilization of preformed Abeta aggregates and attenuation of the cytotoxicity of Abeta oligomers and fibrils could be valuable therapeutics of patients with AD. Recent studies suggested that moderate consumption of red wine and intake of dietary polyphenols, such as resveratrol, may benefit AD phenotypes in animal models and reduce the relative risk for AD clinical dementia. To understand the mechanism of this neuroprotection, we studied the effects of resveratrol, an active ingredient of polyphenols in wine and many plants, on the polymerization of Abeta42 monomer, the destabilization of Abeta42 fibril and the cell toxicity of Abeta42 in vitro using fluorescence spectroscopic analysis with thioflavin T (ThT), transmission electron microscope (TEM), circular dichroism (CD) and MTT assay. The results showed that resveratrol could dose-dependently inhibit Abeta42 fibril formation and cytotoxicity but could not prevent Abeta42 oligomerization. The studies by Western-blot, dot-blot and ELISA confirmed that the addition of resveratrol resulted in numerous Abeta42 oligomer formation. In conjunction with the concept that Abeta oligomers are linked to Abeta toxicity, we speculate that aside from potential antioxidant activities, resveratrol may directly bind to Abeta42, interfere in Abeta42 aggregation, change the Abeta42 oligomer conformation and attenuate Abeta42 oligomeric cytotoxicity.


Asunto(s)
Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Amiloide/efectos de los fármacos , Antioxidantes/farmacología , Estilbenos/farmacología , Amiloide/química , Amiloide/ultraestructura , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/ultraestructura , Animales , Benzotiazoles , Proliferación Celular/efectos de los fármacos , Dicroismo Circular/métodos , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Microscopía Electrónica de Transmisión/métodos , Conformación Molecular , Estructura Molecular , Neuroblastoma/patología , Fragmentos de Péptidos/toxicidad , Estructura Cuaternaria de Proteína , Resveratrol , Espectrometría de Fluorescencia/métodos , Sales de Tetrazolio , Tiazoles/metabolismo
18.
FEBS Lett ; 583(3): 579-84, 2009 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19162022

RESUMEN

Increasing evidence indicates that beta-amyloid (Abeta) oligomers rather than monomers or fibrils are the major toxic agents that specifically inhibit synaptic plasticity and long-term potentiation (LTP) in Alzheimer's disease (AD). Neutralization of Abeta oligomeric toxicity was found to reverse memory deficits. Here, we report four single-chain variable fragment (scFv) antibodies isolated from the naive human scFv library by phage display that specifically recognized Abeta oligomers but not monomers and fibrils. These conformation-dependent scFv antibodies inhibit both Abeta fibrillation and cytotoxicity and bind to the same type of eptitope displayed on the Abeta oligomers. Such scFv antibodies specifically targeting toxic Abeta oligomers may have potential therapeutic and diagnostic applications for AD.


Asunto(s)
Amiloide/inmunología , Amiloide/metabolismo , Anticuerpos/inmunología , Especificidad de Anticuerpos/inmunología , Multimerización de Proteína , Amiloide/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Epítopos/inmunología , Humanos , Cinética , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA