Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 37(17): 4584-4592, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28363982

RESUMEN

Transforming growth factor-ß (TGF-ß) plays an important role in the development and maintenance of embryonic dopaminergic (DA) neurons in the midbrain. To study the function of TGF-ß signaling in the adult nigrostriatal system, we generated transgenic mice with reduced TGF-ß signaling in mature neurons. These mice display age-related motor deficits and degeneration of the nigrostriatal system. Increasing TGF-ß signaling in the substantia nigra through adeno-associated virus expressing a constitutively active type I receptor significantly reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration and motor deficits. These results suggest that TGF-ß signaling is critical for adult DA neuron survival and that modulating this signaling pathway has therapeutic potential in Parkinson disease.SIGNIFICANCE STATEMENT We show that reducing Transforming growth factor-ß (TGF-ß) signaling promotes Parkinson disease-related pathologies and motor deficits, and increasing TGF-ß signaling reduces neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, a parkinsonism-inducing agent. Our results provide a rationale to pursue a means of increasing TGF-ß signaling as a potential therapy for Parkinson's disease.


Asunto(s)
Intoxicación por MPTP/fisiopatología , Neostriado/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Transducción de Señal , Sustancia Negra/fisiopatología , Factor de Crecimiento Transformador beta/deficiencia , Animales , Supervivencia Celular/genética , Trastornos Neurológicos de la Marcha/inducido químicamente , Trastornos Neurológicos de la Marcha/fisiopatología , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/inducido químicamente , Equilibrio Postural , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética
2.
J Alzheimers Dis ; 91(1): 305-319, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36404546

RESUMEN

BACKGROUND: Alzheimer's disease cerebrospinal fluid (CSF) biomarkers amyloid-ß 1-42 (Aß42), total tau (T-tau), and phosphorylated tau 181 (P-tau181) are widely used. However, concentration gradient of these biomarkers between intraventricular (V-CSF) and lumbar CSF (L-CSF) has been demonstrated in idiopathic normal pressure hydrocephalus (iNPH), potentially affecting clinical utility. OBJECTIVE: Here we aim to provide conversion factors for clinical and research use between V-CSF and L-CSF. METHODS: Altogether 138 iNPH patients participated. L-CSF samples were obtained prior to shunt surgery. Intraoperative V-CSF samples were obtained from 97 patients. Post-operative follow-up L- and V-CSF (shunt reservoir) samples of 41 patients were obtained 1-73 months after surgery and then after 3, 6, and 18 months. CSF concentrations of Aß42, T-tau, and P-tau181 were analyzed using commercial ELISA assays. RESULTS: Preoperative L-CSF Aß42, T-tau, and P-tau181 correlated to intraoperative V-CSF (ρ= 0.34-0.55, p < 0.001). Strong correlations were seen between postoperative L- and V-CSF for all biomarkers in every follow-up sampling point (ρs Aß42: 0.77-0.88, T-tau: 0.91-0.94, P-tau181: 0.94-0.96, p < 0.0001). Regression equations were determined for intraoperative V- and preoperative L-CSF (Aß42: V-CSF = 185+0.34*L-CSF, T-tau: Ln(V-CSF) = 3.11+0.49*Ln(L-CSF), P-tau181: V-CSF = 8.2+0.51*L-CSF), and for postoperative V- and L-CSF (Aß42: V-CSF = 86.7+0.75*L-CSF, T-tau: V-CSF = 86.9+0.62*L-CSF, P-tau181: V-CSF = 2.6+0.74*L-CSF). CONCLUSION: Aß42, T-tau, and P-tau181 correlate linearly in-between V- and L-CSF, even stronger after CSF shunt surgery. Equations presented here, provide a novel tool to use V-CSF for diagnostic and prognostic entities relying on the L-CSF concentrations and can be applicable to clinical use when L-CSF samples are not available or less invasively obtained shunt reservoir samples should be interpreted.


Asunto(s)
Enfermedad de Alzheimer , Hidrocéfalo Normotenso , Humanos , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/líquido cefalorraquídeo , Hidrocéfalo Normotenso/líquido cefalorraquídeo , Proteínas tau/líquido cefalorraquídeo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Biomarcadores/líquido cefalorraquídeo , Fragmentos de Péptidos/líquido cefalorraquídeo
3.
J Biol Chem ; 286(47): 40443-51, 2011 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-21956108

RESUMEN

Prodomains of A disintegrin and metalloproteinase (ADAM) metallopeptidases can act as highly specific intra- and intermolecular inhibitors of ADAM catalytic activity. The mouse ADAM9 prodomain (proA9; amino acids 24-204), expressed and characterized from Escherichia coli, is a competitive inhibitor of human ADAM9 catalytic/disintegrin domain with an overall inhibition constant of 280 ± 34 nM and high specificity toward ADAM9. In SY5Y neuroblastoma cells overexpressing amyloid precursor protein, proA9 treatment reduces the amount of endogenous ADAM10 enzyme in the medium while increasing membrane-bound ADAM10, as shown both by Western and activity assays with selective fluorescent peptide substrates using proteolytic activity matrix analysis. An increase in membrane-bound ADAM10 generates higher levels of soluble amyloid precursor protein α in the medium, whereas soluble amyloid precursor protein ß levels are decreased, demonstrating that inhibition of ADAM9 increases α-secretase activity on the cell membrane. Quantification of physiological ADAM10 substrates by a proteomic approach revealed that substrates, such as epidermal growth factor (EGF), HER2, osteoactivin, and CD40-ligand, are increased in the medium of BT474 breast tumor cells that were incubated with proA9, demonstrating that the regulation of ADAM10 by ADAM9 applies for many ADAM10 substrates. Taken together, our results demonstrate that ADAM10 activity is regulated by inhibition of ADAM9, and this regulation may be used to control shedding of amyloid precursor protein by enhancing α-secretase activity, a key regulatory step in the etiology of Alzheimer disease.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas ADAM/química , Proteína ADAM10 , Biocatálisis/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Proteínas de la Membrana/química , Inhibidores de Proteasas/farmacología , Análisis por Matrices de Proteínas , Estructura Terciaria de Proteína
4.
J Neurosci ; 30(14): 4833-44, 2010 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-20371803

RESUMEN

The metalloproteinase and major amyloid precursor protein (APP) alpha-secretase candidate ADAM10 is responsible for the shedding of proteins important for brain development, such as cadherins, ephrins, and Notch receptors. Adam10(-/-) mice die at embryonic day 9.5, due to major defects in development of somites and vasculogenesis. To investigate the function of ADAM10 in brain, we generated Adam10 conditional knock-out (cKO) mice using a Nestin-Cre promotor, limiting ADAM10 inactivation to neural progenitor cells (NPCs) and NPC-derived neurons and glial cells. The cKO mice die perinatally with a disrupted neocortex and a severely reduced ganglionic eminence, due to precocious neuronal differentiation resulting in an early depletion of progenitor cells. Premature neuronal differentiation is associated with aberrant neuronal migration and a disorganized laminar architecture in the neocortex. Neurospheres derived from Adam10 cKO mice have a disrupted sphere organization and segregated more neurons at the expense of astrocytes. We found that Notch-1 processing was affected, leading to downregulation of several Notch-regulated genes in Adam10 cKO brains, in accordance with the central role of ADAM10 in this signaling pathway and explaining the neurogenic phenotype. Finally, we found that alpha-secretase-mediated processing of APP was largely reduced in these neurons, demonstrating that ADAM10 represents the most important APP alpha-secretase in brain. Our study reveals that ADAM10 plays a central role in the developing brain by controlling mainly Notch-dependent pathways but likely also by reducing surface shedding of other neuronal membrane proteins including APP.


Asunto(s)
Proteínas ADAM/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Proteínas de la Membrana/fisiología , Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/deficiencia , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Animales Recién Nacidos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Corteza Cerebral/crecimiento & desarrollo , Femenino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neurogénesis/genética , Neurogénesis/fisiología , Embarazo , Receptores Notch/biosíntesis , Receptores Notch/metabolismo
5.
J Alzheimers Dis ; 80(4): 1629-1642, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33720890

RESUMEN

BACKGROUND: Longitudinal changes in cerebrospinal fluid (CSF) biomarkers are seldom studied. Furthermore, data on biomarker gradient between lumbar (L-) and ventricular (V-) compartments seems to be discordant. OBJECTIVE: To examine alteration of CSF biomarkers reflecting Alzheimer's disease (AD)-related amyloid-ß (Aß) aggregation, tau pathology, neurodegeneration, and early synaptic degeneration by CSF shunt surgery in idiopathic normal pressure hydrocephalus (iNPH) in relation to AD-related changes in brain biopsy. In addition, biomarker levels in L- and V-CSF were compared. METHODS: L-CSF was collected prior to shunt placement and, together with V-CSF, 3-73 months after surgery. Thereafter, additional CSF sampling took place at 3, 6, and 18 months after the baseline sample from 26 iNPH patients with confirmed Aß plaques in frontal cortical brain biopsy and 13 iNPH patients without Aß pathology. CSF Amyloid-ß42 (Aß42), total tau (T-tau), phosphorylated tau (P-tau181), neurofilament light (NFL), and neurogranin (NRGN) were analyzed with customized ELISAs. RESULTS: All biomarkers but Aß42 increased notably by 140-810% in L-CSF after CSF diversion and then stabilized. Aß42 instead showed divergent longitudinal decrease between Aß-positive and -negative patients in L-CSF, and thereafter increase in Aß-negative iNPH patients in both L- and V-CSF. All five biomarkers correlated highly between V-CSF and L-CSF (Aß42 R = 0.87, T-tau R = 0.83, P-tau R = 0.92, NFL R = 0.94, NRGN R = 0.9; all p < 0.0001) but were systematically lower in V-CSF (Aß42 14 %, T-tau 22%, P-tau 20%, NFL 32%, NRGN 19%). With APOE genotype-grouping, only Aß42 showed higher concentration in non-carriers of allele ɛ4. CONCLUSION: Longitudinal follow up shows that after an initial post-surgery increase, T-tau, P-tau, and NRGN are stable in iNPH patients regardless of brain biopsy Aß pathology, while NFL normalized toward its pre-shunt levels. Aß42 as biomarker seems to be the least affected by the surgical procedure or shunt and may be the best predictor of AD risk in iNPH patients. All biomarker concentrations were lower in V- than L-CSF yet showing strong correlations.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Hidrocéfalo Normotenso/líquido cefalorraquídeo , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Fragmentos de Péptidos/líquido cefalorraquídeo , Proteínas tau/líquido cefalorraquídeo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Biomarcadores/líquido cefalorraquídeo , Derivaciones del Líquido Cefalorraquídeo , Femenino , Humanos , Hidrocéfalo Normotenso/patología , Hidrocéfalo Normotenso/cirugía , Masculino , Persona de Mediana Edad , Fosforilación , Análisis de Regresión , Medición de Riesgo
6.
Alzheimers Res Ther ; 12(1): 58, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32410694

RESUMEN

BACKGROUND: Atabecestat, a potent brain-penetrable inhibitor of BACE1 activity that reduces CSF amyloid beta (Aß), was developed for oral treatment for Alzheimer's disease (AD). The long-term safety and effect of atabecestat on cognitive performance in participants with predementia AD in two phase 2 studies were assessed. METHODS: In the placebo-controlled double-blind parent ALZ2002 study, participants aged 50 to 85 years were randomized (1:1:1) to placebo or atabecestat 10 or 50 mg once daily (later reduced to 5 and 25 mg) for 6 months. Participants entered ALZ2004, a 12-month treatment extension with placebo or atabecestat 10 or 25 mg, followed by an open-label phase. Safety, changes in CSF biomarker levels, brain volume, and effects on cognitive performance were assessed. RESULTS: Of 114 participants randomized in ALZ2002, 99 (87%) completed, 90 entered the ALZ2004 double-blind phase, and 77 progressed to the open-label phase. CSF Aß fragments and sAPPß were reduced dose-proportionately. Decreases in whole brain and hippocampal volumes were greater in participants with mild cognitive impairment (MCI) due to AD than in preclinical AD, but were not affected by treatment. In ALZ2004, change from baseline in RBANS trended toward worse scores for atabecestat versus placebo. Elevated liver enzyme adverse events reported in 12 participants on atabecestat resulted in dosage modification and increased frequency of safety monitoring. Treatment discontinuation normalized ALT or AST in all except one with pretreatment elevation, which remained mildly elevated. No case met ALT/AST > 3× ULN and total bilirubin > 2× ULN (Hy's law). CONCLUSION: Atabecestat was associated with trend toward declines in cognition, and elevation of liver enzymes. TRIAL REGISTRATION: ALZ2002: ClinicalTrials.gov, NCT02260674, registered October 9, 2014; ALZ2004: ClinicalTrials.gov, NCT02406027, registered April 1, 2015.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides , Ácido Aspártico Endopeptidasas , Método Doble Ciego , Humanos , Piridinas , Tiazinas , Resultado del Tratamiento
7.
J Neurochem ; 110(4): 1254-62, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19549280

RESUMEN

Transforming growth factor-beta1 (TGF-beta1) has central functions in development, tissue maintenance, and repair and has been implicated in major diseases. We discovered that TGF-beta1 contains several amphipathic helices and hydrophobic domains similar to apolipoprotein E (apoE), a protein involved in lipoprotein metabolism. Indeed, TGF-beta1 associates with lipoproteins isolated from human plasma, cultured liver cells, or astrocytes, and its bioactivity was highest in high-density lipoprotein preparations. Importantly, lipoproteins containing the apoE3 isoform had higher TGF-beta levels and bioactivity than those containing apoE4, a major genetic risk factor for atherosclerosis and Alzheimer's disease. Because TGF-beta1 can be protective in these diseases an association with apoE3 may be beneficial. Association of TGF-beta with different types of lipoproteins may facilitate its diffusion, regulate signaling, and offer additional specificity for this important growth factor.


Asunto(s)
Apolipoproteína E3/metabolismo , Astrocitos/metabolismo , Hepatocitos/metabolismo , Lipoproteínas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Apolipoproteína E3/genética , Apolipoproteína E4/metabolismo , Línea Celular , Células Cultivadas , Humanos , Metabolismo de los Lípidos/fisiología , Ratones , Ratones Noqueados , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína/fisiología , Estructura Terciaria de Proteína/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/química , Factor de Crecimiento Transformador beta1/genética
8.
J Clin Invest ; 116(11): 3060-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17080199

RESUMEN

Alzheimer's disease (AD) is characterized by progressive neurodegeneration and cerebral accumulation of the beta-amyloid peptide (Abeta), but it is unknown what makes neurons susceptible to degeneration. We report that the TGF-beta type II receptor (TbetaRII) is mainly expressed by neurons, and that TbetaRII levels are reduced in human AD brain and correlate with pathological hallmarks of the disease. Reducing neuronal TGF-beta signaling in mice resulted in age-dependent neurodegeneration and promoted Abeta accumulation and dendritic loss in a mouse model of AD. In cultured cells, reduced TGF-beta signaling caused neuronal degeneration and resulted in increased levels of secreted Abeta and beta-secretase-cleaved soluble amyloid precursor protein. These results show that reduced neuronal TGF-beta signaling increases age-dependent neurodegeneration and AD-like disease in vivo. Increasing neuronal TGF-beta signaling may thus reduce neurodegeneration and be beneficial in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Envejecimiento/fisiología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Células Cultivadas , Dendritas/metabolismo , Dendritas/patología , Gliosis/metabolismo , Gliosis/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Serina-Treonina Quinasas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo
9.
Neurobiol Aging ; 79: 131-141, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31055223

RESUMEN

Amyloid ß (Aß) and tau are key hallmark features of Alzheimer's disease (AD) neuropathology. The interplay of Aß and tau for cognitive impairment in early AD was examined with cross-sectional analysis, measured by cerebrospinal fluid biomarkers (Aß1-42, total tau [t-tau], and phosphorylated tau [p-tau181P]), and on cognitive performance by the repeatable battery for assessment of neuropsychological status (RBANS). Participants (n = 246) included cognitively normal (Aß-), mild cognitively impaired (Aß-), preclinical AD (Aß+), and prodromal AD (Aß+). Overall, cognitive scores (RBANS total scale score) had a moderate negative correlation to t-tau (n = 246; r = -0.434; p < 0.001) and p-tau181P (r = -0.389; p < 0.001). When classified by Aß status, this correlation to t-tau was applicable only in Aß+ participants (n = 139; r = -0.451, p < 0.001) but not Aß- participants (n = 107; r = 0.137, p = 0.16), with identical findings for p-tau. Both tau (p < 0.0001) and interaction of Aß1-42 with tau (p = 0.006) affected RBANS, but not Aß1-42 alone. Cognitive/memory performance correlated well with cerebrospinal fluid tau levels across early stages of AD, although the correlation is Aß dependent.


Asunto(s)
Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/líquido cefalorraquídeo , Cognición , Fragmentos de Péptidos/líquido cefalorraquídeo , Proteínas tau/líquido cefalorraquídeo , Anciano , Enfermedad de Alzheimer/diagnóstico , Biomarcadores/líquido cefalorraquídeo , Estudios Transversales , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas
10.
Neuron ; 40(6): 1133-45, 2003 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-14687548

RESUMEN

TGF-beta1 is a key regulator of diverse biological processes in many tissues and cell types, but its exact function in the developing and adult mammalian CNS is still unknown. We report that lack of TGF-beta1 expression in neonatal Tgfb1(-/-) mice results in a widespread increase in degenerating neurons accompanied by reduced expression of synaptophysin and laminin and a prominent microgliosis. Lack of TGF-beta1 also strongly reduces survival of primary neurons cultured from Tgfb1(-/-) mice. TGF-beta1 deficiency in adult Tgfb1(-/+) mice results in increased neuronal susceptibility to excitotoxic injury, whereas astroglial overexpression of TGF-beta1 protects adult mice against neurodegeneration in acute, excitotoxic and chronic injury paradigms. This study reveals a nonredundant function for TGF-beta1 in maintaining neuronal integrity and survival of CNS neurons and in regulating microglial activation. Because individual TGF-beta1 expression levels in the brain vary considerably between humans, this finding could have important implications for susceptibility to neurodegeneration.


Asunto(s)
Encéfalo/metabolismo , Gliosis/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Factor de Crecimiento Transformador beta/deficiencia , Animales , Encéfalo/patología , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Gliosis/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Neuronas/patología , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1
11.
Alzheimers Res Ther ; 10(1): 85, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30134967

RESUMEN

BACKGROUND: ß-Secretase enzyme (BACE) inhibition has been proposed as a priority treatment mechanism for Alzheimer's disease (AD), but treatment initiation may need to be very early. We present proof of mechanism of atabecestat (also known as JNJ-54861911), an oral BACE inhibitor for the treatment of AD, in Caucasian and Japanese populations with early AD who do not show signs of dementia. METHODS: In two similarly designed phase I studies, a sample of amyloid-positive elderly patients comprising 45 Caucasian patients with early AD diagnosed as preclinical AD (n = 15, Clinical Dementia Rating [CDR] = 0) or with mild cognitive impairment due to AD (n = 30, CDR = 0.5) and 18 Japanese patients diagnosed as preclinical AD (CDR-J = 0) were randomized 1:1:1 to atabecestat 10 or 50 mg or placebo (n = 6-8/treatment) daily for 4 weeks. Safety, pharmacokinetics (PK), and pharmacodynamics (PD) (i.e., reduction of cerebrospinal fluid [CSF] amyloid beta 1-40 [Aß1-40] levels [primary endpoint] and effect on other AD biomarkers) of atabecestat were evaluated. RESULTS: In both populations, atabecestat was well tolerated and characterized by linear PK and high central nervous system penetrance of unbound drug. Atabecestat significantly reduced CSF Aß1-40 levels from baseline at day 28 in both the 10-mg (67-68%) and 50-mg (87-90%) dose groups compared with placebo. For Caucasians with early AD, the least squares mean differences (95% CI) were - 69.37 (- 72.25; - 61.50) and - 92.74 (- 100.08; - 85.39), and for Japanese with preclinical AD, they were - 62.48 (- 78.32; - 46.64) and - 80.81 (- 96.13; - 65.49), respectively. PK/PD model simulations confirmed that once-daily 10 mg and 50 mg atabecestat can attain 60-70% and 90% Aß1-40 reductions, respectively. The trend of the reduction was similar across the Aß1-37, Aß1-38, and Aß1-42 fragments in both atabecestat dose groups, consistent with Aß1-40. CSF amyloid precursor protein fragment (sAPPß) levels declined from baseline, regardless of patient population, whereas CSF sAPPα levels increased compared with placebo. There were no relevant changes in either CSF total tau or phosphorylated tau 181P over a 4-week treatment period. CONCLUSIONS: JNJ-54861911 at 10 and 50 mg daily doses after 4 weeks resulted in mean CSF Aß1-40 reductions of 67% and up to 90% in both Caucasian and Japanese patients with early stage AD, confirming results in healthy elderly adults. TRIAL REGISTRATION: ALZ1005: ClinicalTrials.gov, NCT01978548. Registered on 7 November 2013. ALZ1008: ClinicalTrials.gov, NCT02360657. Registered on 10 February 2015.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/líquido cefalorraquídeo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Fragmentos de Péptidos/líquido cefalorraquídeo , Piridinas/farmacología , Tiazinas/farmacología , Administración Oral , Anciano , Enfermedad de Alzheimer/líquido cefalorraquídeo , Pueblo Asiatico , Biomarcadores/líquido cefalorraquídeo , Método Doble Ciego , Femenino , Humanos , Masculino , Piridinas/administración & dosificación , Tiazinas/administración & dosificación , Resultado del Tratamiento , Población Blanca , Proteínas tau/líquido cefalorraquídeo
12.
J Alzheimers Dis ; 64(1): 171-179, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29865068

RESUMEN

BACKGROUND: Detection of pathological tau aggregates could facilitate clinical diagnosis of Alzheimer's disease (AD) and monitor drug effects in clinical trials. S-[18F]THK-5117 could be a potential tracer to detect pathological tau deposits in brain. However, no previous study have correlated S-[18F]THK-5117 uptake in PET with brain biopsy verified tau pathology in vivo. OBJECTIVE: Here we aim to evaluate the association between cerebrospinal fluid (CSF) AD biomarkers, S-[18F]THK-5117, and [11C]PIB PET against tau and amyloid lesions in brain biopsy. METHODS: Fourteen patients with idiopathic normal pressure hydrocephalus (iNPH) with previous shunt surgery including right frontal cortical brain biopsy and CSF Aß1 - 42, total tau, and P-tau181 measures, underwent brain MRI, [11C]PIB PET, and S-[18F]THK-5117 PET imaging. RESULTS: Seven patients had amyloid-ß (Aß, 4G8) plaques, two both Aß and phosphorylated tau (Pτ, AT8) and one only Pτ in biopsy. As expected, increased brain biopsy Aß was well associated with higher [11C]PIB uptake in PET. However, S-[18F]THK-5117 uptake did not show any statistically significant correlation with either brain biopsy Pτ or CSF P-tau181 or total tau. CONCLUSIONS: S-[18F]THK-5117 lacked clear association with neuropathologically verified tau pathology in brain biopsy probably, at least partially, due to off-target binding. Further studies with larger samples of patients with different tau tracers are urgently needed. The detection of simultaneous Aß and tau pathology in iNPH is important since that may indicate poorer and especially shorter response for CSF shunt surgery compared with no pathology.


Asunto(s)
Compuestos de Anilina/farmacocinética , Encéfalo , Hidrocéfalo Normotenso/diagnóstico por imagen , Placa Amiloide/metabolismo , Quinolinas/farmacocinética , Tiazoles/farmacocinética , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Mapeo Encefálico , Femenino , Humanos , Hidrocéfalo Normotenso/líquido cefalorraquídeo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones
13.
J Alzheimers Dis ; 56(4): 1437-1449, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28157093

RESUMEN

The ß-site amyloid-ß protein precursor (AßPP) cleaving enzyme-1 (BACE1) is the rate limiting enzyme in the generation of amyloid-ß peptide (Aß) from AßPP, one of the major pathways in Alzheimer's disease (AD) pathology. Increased BACE1 levels and activity have been reported in the brain of patients with sporadic AD. Therefore, changes of BACE1 levels in the cerebrospinal fluid (CSF) have also been investigated as a possible biomarker of the disease. We analyzed BACE1 levels in CSF of elderly healthy participants before and after chronic treatment with a BACE inhibitor (BACEi) and evaluated the correlation between BACE1 levels and downstream AD markers. Overall, BACE1 CSF levels showed strong correlations to all downstream AD markers investigated. This is the first reported finding that shows BACE1 levels in CSF were well correlated to its end product Aß1 - 42. As previously described, BACE1 levels were strongly correlated to total-tau and phosphorylated tau levels in CSF. Generally, chronic BACE inhibition did not influence BACE1 CSF protein levels. Follow-up studies including early-stage AD pathophysiology and prodromal AD patients will help to understand the importance of measuring BACE1 routinely in daily clinical practice and AD clinical trials.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Inhibidores de Proteasas/uso terapéutico , Proteínas tau/líquido cefalorraquídeo , Anciano , Enfermedad de Alzheimer/líquido cefalorraquídeo , Apolipoproteína E4/genética , Biomarcadores/líquido cefalorraquídeo , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Inhibidores de Proteasas/efectos adversos , Inhibidores de Proteasas/farmacocinética
14.
Neuron ; 93(5): 1066-1081.e8, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28238547

RESUMEN

Human pluripotent stem cells (PSCs) provide a unique entry to study species-specific aspects of human disorders such as Alzheimer's disease (AD). However, in vitro culture of neurons deprives them of their natural environment. Here we transplanted human PSC-derived cortical neuronal precursors into the brain of a murine AD model. Human neurons differentiate and integrate into the brain, express 3R/4R Tau splice forms, show abnormal phosphorylation and conformational Tau changes, and undergo neurodegeneration. Remarkably, cell death was dissociated from tangle formation in this natural 3D model of AD. Using genome-wide expression analysis, we observed upregulation of genes involved in myelination and downregulation of genes related to memory and cognition, synaptic transmission, and neuron projection. This novel chimeric model for AD displays human-specific pathological features and allows the analysis of different genetic backgrounds and mutations during the course of the disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo , Diferenciación Celular/fisiología , Neuritas/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/diagnóstico , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular/fisiología , Humanos , Ratones , Fosforilación
15.
BMC Cell Biol ; 7: 15, 2006 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-16549026

RESUMEN

BACKGROUND: Transforming Growth Factor-beta (TGF-beta) regulates key biological processes during development and in adult tissues and has been implicated in many diseases. To study the biological functions of TGF-beta, sensitive, specific, and convenient bioassays are necessary. Here we describe a new cell-based bioassay that fulfills these requirements. RESULTS: Embryonic fibroblasts from Tgfb1-/- mice were stably transfected with a reporter plasmid consisting of TGF-beta responsive Smad-binding elements coupled to a secreted alkaline phosphatase reporter gene (SBE-SEAP). Clone MFB-F11 showed more than 1000-fold induction after stimulation with 1 ng/ml TGF-beta1, and detected as little as 1 pg/ml TGF-beta1. MFB-F11 cells were highly induced by TGF-beta1, TGF-beta2 and TGF-beta3, but did not show induction with related family members activin, nodal, BMP-2 and BMP-6 or with trophic factors bFGF and BDNF. MFB-F11 cells can detect and quantify TGF-beta in biological samples without prior enrichment of TGF-betas, and can detect biologically activated TGF-beta in a cell co-culture system. CONCLUSION: MFB-F11 cells can be used to rapidly and specifically measure TGF-beta with high sensitivity.


Asunto(s)
Bioensayo/métodos , Factor de Crecimiento Transformador beta/análisis , Fosfatasa Alcalina/genética , Animales , Química Encefálica , Células CHO , Línea Celular , Técnicas de Cocultivo/métodos , Cricetinae , Medios de Cultivo/química , Femenino , Fibroblastos/química , Fibroblastos/citología , Genes Reporteros/genética , Ratones , Ratones Mutantes , Sensibilidad y Especificidad , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/sangre , Factor de Crecimiento Transformador beta/genética
16.
Curr Alzheimer Res ; 3(5): 505-13, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17168649

RESUMEN

Neurodegenerative diseases including Alzheimer's disease (AD) and Parkinson's disease (PD) afflict growing numbers of people but treatments are not available or ineffective. These diseases are characterized by the loss of specific neuronal populations, the accumulation of protein aggregates inside and sometimes outside neurons, and an activation of immune pathways in the brain. The causes of sporadic forms of AD or PD are not known but it has been postulated that reduced trophic support to neurons together with age dependent increases in cellular stress lead to chronic injury and ultimately the demise of neurons. TGF-betas are neuroprotective factors and organizers of injury responses and as such might have a role in neurodegenerative disease. We review here the evidence mostly from genetically manipulated mice that links the TGF-beta signaling pathway to neuronal phenotypes and neurodegeneration. Although many of these mutant models did not produce overt CNS phenotypes or adult brain were not studied due to embryonic lethality, there is growing support for a role of TGF-beta signaling in neuronal maintenance, function, and degeneration. Future studies will have to determine whether dysregulation of TGF-beta signaling in neurodegenerative diseases is significant and whether this signaling pathway may even be a target for treatment.


Asunto(s)
Animales Modificados Genéticamente , Enfermedades Neurodegenerativas/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
17.
J Neurosci ; 24(10): 2527-34, 2004 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-15014128

RESUMEN

Apolipoprotein E (apoE) is found in amyloid plaques and neurofibrillary tangles (NFTs) in Alzheimer's disease (AD) brains, but its role in their pathogenesis is unclear. Previously, we found C-terminal-truncated fragments of apoE in AD brains and showed that such fragments can cause neurodegeneration and can induce NFT-like inclusions in cultured neuronal cells and in transgenic mice. Here, we analyzed apoE fragmentation in brain tissue homogenates from transgenic mice expressing apoE3 or apoE4 in neurons [neuron-specific enolase (NSE)-apoE] or astrocytes [glial fibrillary acidic protein (GFAP)-apoE] by Western blotting. The C-terminal-truncated fragments of apoE accumulated, in an age-dependent manner, in the brains of NSE-apoE4 and, to a significantly lesser extent, NSE-apoE3 mice; however, no fragments were detected in GFAP-apoE3 or GFAP-apoE4 mice. In NSE-apoE mice, the pattern of apoE fragmentation resembled that seen in AD brains, and the fragmentation was specific for certain brain regions, occurring in the neocortex and hippocampus, which are vulnerable to AD-related neurodegeneration, but not in the less vulnerable cerebellum. Excitotoxic challenge with kainic acid significantly increased apoE fragmentation in NSE-apoE4 but not NSE-apoE3 mice. Phosphorylated tau (p-tau) also accumulated in an age-dependent manner in NSE-apoE4 mice and, to a much lesser extent, in NSE-apoE3 mice but not in GFAP-apoE3 or GFAP-apoE4 mice. Intraneuronal p-tau inclusions in the hippocampus were prominent in 21-month-old NSE-apoE4 mice but barely detectable in NSE-apoE3 mice. Thus, the accumulation of potentially pathogenic C-terminal-truncated fragments of apoE depends on both the isoform and the cellular source of apoE. Neuron-specific proteolytic cleavage of apoE4 is associated with increased phosphorylation of tau and may play a key role in the development of AD-related neuronal deficits.


Asunto(s)
Enfermedad de Alzheimer/genética , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Envejecimiento/metabolismo , Animales , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/genética , Astrocitos/citología , Astrocitos/metabolismo , Encéfalo/citología , Encéfalo/efectos de los fármacos , Química Encefálica/genética , Femenino , Marcación de Gen , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Ácido Kaínico/farmacología , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Neurotoxinas/farmacología , Fragmentos de Péptidos/metabolismo , Fosfopiruvato Hidratasa/genética , Fosforilación , Regiones Promotoras Genéticas
18.
Curr Alzheimer Res ; 2(2): 183-6, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15974916

RESUMEN

Neurodegenerative and dementing illnesses are becoming an increasing social and economical burden as the number of older people continues to grow in industrialized countries. Current knowledge of the processes leading to these diseases is still limited, and very few effective treatments are available. Because neurodegeneration is associated with an activation of injury and innate immune responses in the brain, drugs that could mimic the beneficial aspects of this response are potential therapeutic candidates. The cytokine transforming growth factor (TGF)-beta1 is an organizer of the brain's response to injury and is known to be neuroprotective. Previous studies from our lab also showed that TGF-beta1 can reduce accumulation of beta-amyloid peptide (Abeta), which appears to be central to Alzheimer's disease (AD) pathogenesis, and we therefore initiated a search for small molecule chemical compounds that could mimic this effect. We report here the identification of several such TGF-beta mimetics detected in an in vitro screen of a library with 5000 chemically diverse compounds. If active in vivo, these mimetics could be developed into candidates for the treatment of neurodegeneration.


Asunto(s)
Materiales Biomiméticos/farmacología , Fármacos Neuroprotectores/farmacología , Factor de Crecimiento Transformador beta/farmacología , Enfermedad de Alzheimer/tratamiento farmacológico , Animales , Materiales Biomiméticos/uso terapéutico , Línea Celular , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Ratones , Ratones Noqueados , Fármacos Neuroprotectores/uso terapéutico , Ratas , Factor de Crecimiento Transformador beta/uso terapéutico
19.
Methods Mol Biol ; 1187: 223-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25053493

RESUMEN

With the increasing appreciation of the role of Notch in development and disease, measuring its cleavage and signaling activity in cellular systems has become important. Here we describe a cell-based method to analyze the cleavage of Notch at the S3 site by γ-secretase. HEK cells are transfected with an N-terminal truncated and myc-labeled mNotchΔE construct which can be easily and quantitatively detected by western blotting.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteolisis , Receptores Notch/metabolismo , Animales , Western Blotting/métodos , Células HEK293 , Humanos , Ratones , Receptores Notch/análisis , Receptores Notch/genética , Transfección/métodos
20.
PLoS One ; 9(1): e87014, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24466315

RESUMEN

Proteolytic processing of the amyloid precursor protein (APP) by the ß- and γ-secretases releases the amyloid-ß peptide (Aß), which deposits in senile plaques and contributes to the etiology of Alzheimer's disease (AD). The α-secretase cleaves APP in the Aß peptide sequence to generate soluble APPα (sAPPα). Upregulation of α-secretase activity through the 5-hydroxytryptamine 4 (5-HT4) receptor has been shown to reduce Aß production, amyloid plaque load and to improve cognitive impairment in transgenic mouse models of AD. Consequently, activation of 5-HT4 receptors following agonist stimulation is considered to be a therapeutic strategy for AD treatment; however, the signaling cascade involved in 5-HT4 receptor-stimulated proteolysis of APP remains to be determined. Here we used chemical and siRNA inhibition to identify the proteins which mediate 5-HT4d receptor-stimulated α-secretase activity in the SH-SY5Y human neuronal cell line. We show that G protein and Src dependent activation of phospholipase C are required for α-secretase activity, while, unexpectedly, adenylyl cyclase and cAMP are not involved. Further elucidation of the signaling pathway indicates that inositol triphosphate phosphorylation and casein kinase 2 activation is also a prerequisite for α-secretase activity. Our findings provide a novel route to explore the treatment of AD through 5-HT4 receptor-induced α-secretase activation.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Serotonina/metabolismo , Transducción de Señal/fisiología , Adenilil Ciclasas/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Arrestinas/metabolismo , Quinasa de la Caseína II/metabolismo , Línea Celular Tumoral , AMP Cíclico/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Inositol Polifosfato 5-Fosfatasas , Ratones , Ratones Transgénicos/metabolismo , Ratones Transgénicos/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteolisis , Receptores de Serotonina 5-HT4/metabolismo , Fosfolipasas de Tipo C/metabolismo , beta-Arrestinas , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA