Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 612(7938): 123-131, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36385530

RESUMO

Aggregates of medin amyloid (a fragment of the protein MFG-E8, also known as lactadherin) are found in the vasculature of almost all humans over 50 years of age1,2, making it the most common amyloid currently known. We recently reported that medin also aggregates in blood vessels of ageing wild-type mice, causing cerebrovascular dysfunction3. Here we demonstrate in amyloid-ß precursor protein (APP) transgenic mice and in patients with Alzheimer's disease that medin co-localizes with vascular amyloid-ß deposits, and that in mice, medin deficiency reduces vascular amyloid-ß deposition by half. Moreover, in both the mouse and human brain, MFG-E8 is highly enriched in the vasculature and both MFG-E8 and medin levels increase with the severity of vascular amyloid-ß burden. Additionally, analysing data from 566 individuals in the ROSMAP cohort, we find that patients with Alzheimer's disease have higher MFGE8 expression levels, which are attributable to vascular cells and are associated with increased measures of cognitive decline, independent of plaque and tau pathology. Mechanistically, we demonstrate that medin interacts directly with amyloid-ß to promote its aggregation, as medin forms heterologous fibrils with amyloid-ß, affects amyloid-ß fibril structure, and cross-seeds amyloid-ß aggregation both in vitro and in vivo. Thus, medin could be a therapeutic target for prevention of vascular damage and cognitive decline resulting from amyloid-ß deposition in the blood vessels of the brain.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Precursor de Proteína beta-Amiloide , Animais , Humanos , Camundongos , Pessoa de Meia-Idade , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Disfunção Cognitiva , Camundongos Transgênicos , Placa Amiloide/metabolismo , Proteínas tau/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(24): e2119804119, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35666874

RESUMO

Single-cell transcriptomics has revealed specific glial activation states associated with the pathogenesis of neurodegenerative diseases, such as Alzheimer's and Parkinson's disease. While these findings may eventually lead to new therapeutic opportunities, little is known about how these glial responses are reflected by biomarker changes in bodily fluids. Such knowledge, however, appears crucial for patient stratification, as well as monitoring disease progression and treatment responses in clinical trials. Here, we took advantage of well-described mouse models of ß-amyloidosis and α-synucleinopathy to explore cerebrospinal fluid (CSF) proteome changes related to their respective proteopathic lesions. Nontargeted liquid chromatography-mass spectrometry revealed that the majority of proteins that undergo age-related changes in CSF of either mouse model were linked to microglia and astrocytes. Specifically, we identified a panel of more than 20 glial-derived proteins that were increased in CSF of aged ß-amyloid precursor protein- and α-synuclein-transgenic mice and largely overlap with previously described disease-associated glial genes identified by single-cell transcriptomics. Our results also show that enhanced shedding is responsible for the increase of several of the identified glial CSF proteins as exemplified for TREM2. Notably, the vast majority of these proteins can also be quantified in human CSF and reveal changes in Alzheimer's disease cohorts. The finding that cellular transcriptome changes translate into corresponding changes of CSF proteins is of clinical relevance, supporting efforts to identify fluid biomarkers that reflect the various functional states of glial responses in cerebral proteopathies, such as Alzheimer's and Parkinson's disease.


Assuntos
Doença de Alzheimer , Líquido Cefalorraquidiano , Neuroglia , Doença de Parkinson , Proteoma , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/metabolismo , Animais , Biomarcadores/líquido cefalorraquidiano , Líquido Cefalorraquidiano/metabolismo , Perfilação da Expressão Gênica , Humanos , Camundongos , Neuroglia/metabolismo , Doença de Parkinson/líquido cefalorraquidiano , Doença de Parkinson/metabolismo , Proteoma/metabolismo , Análise de Célula Única , Proteínas tau
3.
EMBO Rep ; 18(9): 1536-1544, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28701326

RESUMO

Little is known about the extent to which pathogenic factors drive the development of Alzheimer's disease (AD) at different stages of the long preclinical and clinical phases. Given that the aggregation of the ß-amyloid peptide (Aß) is an important factor in AD pathogenesis, we asked whether Aß seeds from brain extracts of mice at different stages of amyloid deposition differ in their biological activity. Specifically, we assessed the effect of age on Aß seeding activity in two mouse models of cerebral Aß amyloidosis (APPPS1 and APP23) with different ages of onset and rates of progression of Aß deposition. Brain extracts from these mice were serially diluted and inoculated into host mice. Strikingly, the seeding activity (seeding dose SD50) in extracts from donor mice of both models reached a plateau relatively early in the amyloidogenic process. When normalized to total brain Aß, the resulting specific seeding activity sharply peaked at the initial phase of Aß deposition, which in turn is characterized by a temporary several-fold increase in the Aß42/Aß40 ratio. At all stages, the specific seeding activity of the APPPS1 extract was higher compared to that of APP23 brain extract, consistent with a more important contribution of Aß42 than Aß40 to seed activity. Our findings indicate that the Aß seeding potency is greatest early in the pathogenic cascade and diminishes as Aß increasingly accumulates in brain. The present results provide experimental support for directing anti-Aß therapeutics to the earliest stage of the pathogenic cascade, preferably before the onset of amyloid deposition.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Amiloidose/metabolismo , Encéfalo/metabolismo , Fatores Etários , Doença de Alzheimer/tratamento farmacológico , Amiloidose/tratamento farmacológico , Amiloidose/fisiopatologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Camundongos Transgênicos
4.
J Neurosci ; 34(31): 10264-73, 2014 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-25080588

RESUMO

Deposition of aggregated amyloid-ß (Aß) peptide in brain is an early event and hallmark pathology of Alzheimer's disease and cerebral Aß angiopathy. Experimental evidence supports the concept that Aß multimers can act as seeds and structurally corrupt other Aß peptides by a self-propagating mechanism. Here we compare the induction of cerebral ß-amyloidosis by intraperitoneal applications of Aß-containing brain extracts in three Aß-precursor protein (APP) transgenic mouse lines that differ in levels of transgene expression in brain and periphery (APP23 mice, APP23 mice lacking murine APP, and R1.40 mice). Results revealed that beta-amyloidosis induction, which could be blocked with an anti-Aß antibody, was dependent on the amount of inoculated brain extract and on the level of APP/Aß expression in the brain but not in the periphery. The induced Aß deposits in brain occurred in a characteristic pattern consistent with the entry of Aß seeds at multiple brain locations. Intraperitoneally injected Aß could be detected in blood monocytes and some peripheral tissues (liver, spleen) up to 30 d after the injection but escaped histological and biochemical detection thereafter. These results suggest that intraperitoneally inoculated Aß seeds are transported from the periphery to the brain in which corruptive templating of host Aß occurs at multiple sites, most efficiently in regions with high availability of soluble Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/toxicidade , Amiloidose , Córtex Cerebral/patologia , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/genética , Amiloidose/induzido quimicamente , Amiloidose/genética , Amiloidose/patologia , Animais , Anticorpos/farmacologia , Células Sanguíneas/metabolismo , Células Sanguíneas/patologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Cavidade Peritoneal/patologia , Placa Amiloide/patologia , Fatores de Tempo
5.
Nat Commun ; 13(1): 7333, 2022 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-36443293

RESUMO

Brain Aß deposition is a key early event in the pathogenesis of Alzheimer´s disease (AD), but the long presymptomatic phase and poor correlation between Aß deposition and clinical symptoms remain puzzling. To elucidate the dependency of downstream pathologies on Aß, we analyzed the trajectories of cerebral Aß accumulation, Aß seeding activity, and neurofilament light chain (NfL) in the CSF (a biomarker of neurodegeneration) in Aß-precursor protein transgenic mice. We find that Aß deposition increases linearly until it reaches an apparent plateau at a late age, while Aß seeding activity increases more rapidly and reaches a plateau earlier, coinciding with the onset of a robust increase of CSF NfL. Short-term inhibition of Aß generation in amyloid-laden mice reduced Aß deposition and associated glial changes, but failed to reduce Aß seeding activity, and CSF NfL continued to increase although at a slower pace. When short-term or long-term inhibition of Aß generation was started at pre-amyloid stages, CSF NfL did not increase despite some Aß deposition, microglial activation, and robust brain Aß seeding activity. A dissociation of Aß load and CSF NfL trajectories was also found in familial AD, consistent with the view that Aß aggregation is not kinetically coupled to neurotoxicity. Rather, neurodegeneration starts when Aß seeding activity is saturated and before Aß deposition reaches critical (half-maximal) levels, a phenomenon reminiscent of the two pathogenic phases in prion disease.


Assuntos
Doença de Alzheimer , Amiloidose , Animais , Camundongos , Encéfalo , Progressão da Doença , Proteínas Amiloidogênicas , Inibição Psicológica , Camundongos Transgênicos
6.
Acta Neuropathol Commun ; 8(1): 133, 2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32787922

RESUMO

Alpha-synucleinopathies are a group of progressive neurodegenerative disorders, characterized by intracellular deposits of aggregated α-synuclein (αS). The clinical heterogeneity of these diseases is thought to be attributed to conformers (or strains) of αS but the contribution of inclusions in various cell types is unclear. The aim of the present work was to study αS conformers among different transgenic (TG) mouse models of α-synucleinopathies. To this end, four different TG mouse models were studied (Prnp-h[A53T]αS; Thy1-h[A53T]αS; Thy1-h[A30P]αS; Thy1-mαS) that overexpress human or murine αS and differed in their age-of-symptom onset and subsequent disease progression. Postmortem analysis of end-stage brains revealed robust neuronal αS pathology as evidenced by accumulation of αS serine 129 (p-αS) phosphorylation in the brainstem of all four TG mouse lines. Overall appearance of the pathology was similar and only modest differences were observed among additionally affected brain regions. To study αS conformers in these mice, we used pentameric formyl thiophene acetic acid (pFTAA), a fluorescent dye with amyloid conformation-dependent spectral properties. Unexpectedly, besides the neuronal αS pathology, we also found abundant pFTAA-positive inclusions in microglia of all four TG mouse lines. These microglial inclusions were also positive for Thioflavin S and showed immunoreactivity with antibodies recognizing the N-terminus of αS, but were largely p-αS-negative. In all four lines, spectral pFTAA analysis revealed conformational differences between microglia and neuronal inclusions but not among the different mouse models. Concomitant with neuronal lesions, microglial inclusions were already present at presymptomatic stages and could also be induced by seeded αS aggregation. Although nature and significance of microglial inclusions for human α-synucleinopathies remain to be clarified, the previously overlooked abundance of microglial inclusions in TG mouse models of α-synucleinopathy bears importance for mechanistic and preclinical-translational studies.


Assuntos
Microglia/patologia , Neurônios/patologia , Sinucleinopatias/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Corpos de Inclusão/patologia , Camundongos , Camundongos Transgênicos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/patologia , Conformação Proteica , Sinucleinopatias/genética , alfa-Sinucleína/química
7.
Nat Neurosci ; 23(12): 1580-1588, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33199898

RESUMO

Amyloid-ß (Aß) deposits are a relatively late consequence of Aß aggregation in Alzheimer's disease. When pathogenic Aß seeds begin to form, propagate and spread is not known, nor are they biochemically defined. We tested various antibodies for their ability to neutralize Aß seeds before Aß deposition becomes detectable in Aß precursor protein-transgenic mice. We also characterized the different antibody recognition profiles using immunoprecipitation of size-fractionated, native, mouse and human brain-derived Aß assemblies. At least one antibody, aducanumab, after acute administration at the pre-amyloid stage, led to a significant reduction of Aß deposition and downstream pathologies 6 months later. This demonstrates that therapeutically targetable pathogenic Aß seeds already exist during the lag phase of protein aggregation in the brain. Thus, the preclinical phase of Alzheimer's disease-currently defined as Aß deposition without clinical symptoms-may be a relatively late manifestation of a much earlier pathogenic seed formation and propagation that currently escapes detection in vivo.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/farmacologia , Química Encefálica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteínas de Neurofilamentos/líquido cefalorraquidiano , Placa Amiloide/patologia , Extratos de Tecidos/farmacologia
8.
J Med Chem ; 62(4): 2038-2048, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30707834

RESUMO

A fluorescent bis-styryl-benzothiadiazole (BTD) with carboxylic acid functional groups (X-34/Congo red analogue) showed lower binding affinity toward Aß1-42 and Aß1-40 fibrils than its neutral analogue. Hence, variable patterns of neutral OH-substituted bis-styryl-BTDs were generated. All bis-styryl-BTDs showed higher binding affinity to Aß1-42 fibrils than to Aß1-40 fibrils. The para-OH on the phenyl rings was beneficial for binding affinity while a meta-OH decreased the affinity. Differential staining of transgenic mouse Aß amyloid plaque cores compared to peripheral coronas using neutral compared to anionic bis-styryl ligands indicate differential recognition of amyloid polymorphs. Hyperspectral imaging of transgenic mouse Aß plaque stained with uncharged para-hydroxyl substituted bis-styryl-BTD implicated differences in binding site polarity of polymorphic amyloid plaque. Most properties of the corresponding bis-styryl-BTD were retained with a rigid alkyne linker rendering a probe insensitive to cis-trans isomerization. These new BTD-based ligands are promising probes for spectral imaging of different Aß fibril polymorphs.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Corantes Fluorescentes/farmacologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Estirenos/farmacologia , Tiadiazóis/farmacologia , Animais , Feminino , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Ligantes , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Ligação Proteica , Estirenos/síntese química , Estirenos/metabolismo , Tiadiazóis/síntese química , Tiadiazóis/metabolismo
9.
Nat Neurosci ; 20(10): 1371-1376, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28846081

RESUMO

To clarify the role of microglia in brain homeostasis and disease, an understanding of their maintenance, proliferation and turnover is essential. The lifespan of brain microglia, however, remains uncertain, and reflects confounding factors in earlier assessments that were largely indirect. We genetically labeled single resident microglia in living mice and then used multiphoton microscopy to monitor these cells over time. Under homeostatic conditions, we found that neocortical resident microglia were long-lived, with a median lifetime of well over 15 months; thus, approximately half of these cells survive the entire mouse lifespan. While proliferation of resident neocortical microglia under homeostatic conditions was low, microglial proliferation in a mouse model of Alzheimer's ß-amyloidosis was increased threefold. The persistence of individual microglia throughout the mouse lifespan provides an explanation for how microglial priming early in life can induce lasting functional changes and how microglial senescence may contribute to age-related neurodegenerative diseases.


Assuntos
Envelhecimento/fisiologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Microglia/citologia , Microglia/fisiologia , Análise de Célula Única , Animais , Morte Celular , Proliferação de Células , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Microglia/patologia , Microscopia de Fluorescência por Excitação Multifotônica , Neocórtex/fisiologia , Placa Amiloide/patologia
10.
Sci Rep ; 6: 28125, 2016 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-27311744

RESUMO

An early event in Alzheimer's disease (AD) pathogenesis is the formation of extracellular aggregates of amyloid-ß peptide (Aß), thought to be initiated by a prion-like seeding mechanism. However, the molecular nature and location of the Aß seeds remain rather elusive. Active Aß seeds are found in crude homogenates of amyloid-laden brains and in the soluble fraction thereof. To analyze the seeding activity of the pellet fraction, we have either separated or directly immunoisolated membranes from such homogenates. Here, we found considerable Aß seeding activity associated with membranes in the absence of detectable amyloid fibrils. We also found that Aß seeds on mitochondrial or associated membranes efficiently induced Aß aggregation in vitro and seed ß-amyloidosis in vivo. Aß seeds at intracellular membranes may contribute to the spreading of Aß aggregation along neuronal pathways and to the induction of intracellular pathologies downstream of Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Amiloide/metabolismo , Mitocôndrias/patologia , Membranas Mitocondriais/patologia , Placa Amiloide/patologia , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
11.
Nat Neurosci ; 18(11): 1559-61, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26352792

RESUMO

Cerebral ß-amyloidosis is induced by inoculation of Aß seeds into APP transgenic mice, but not into App(-/-) (APP null) mice. We found that brain extracts from APP null mice that had been inoculated with Aß seeds up to 6 months previously still induced ß-amyloidosis in APP transgenic hosts following secondary transmission. Thus, Aß seeds can persist in the brain for months, and they regain propagative and pathogenic activity in the presence of host Aß.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/administração & dosagem , Precursor de Proteína beta-Amiloide/deficiência , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/patologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placa Amiloide/patologia
12.
EMBO Mol Med ; 7(7): 895-903, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25978969

RESUMO

Abnormalities in brains of Alzheimer's disease (AD) patients are thought to start long before the first clinical symptoms emerge. The identification of affected individuals at this 'preclinical AD' stage relies on biomarkers such as decreased levels of the amyloid-ß peptide (Aß) in the cerebrospinal fluid (CSF) and positive amyloid positron emission tomography scans. However, there is little information on the longitudinal dynamics of CSF biomarkers, especially in the earliest disease stages when therapeutic interventions are likely most effective. To this end, we have studied CSF Aß changes in three Aß precursor protein transgenic mouse models, focusing our analysis on the initial Aß deposition, which differs significantly among the models studied. Remarkably, while we confirmed the CSF Aß decrease during the extended course of brain Aß deposition, a 20-30% increase in CSF Aß40 and Aß42 was found around the time of the first Aß plaque appearance in all models. The biphasic nature of this observed biomarker changes stresses the need for longitudinal biomarker studies in the clinical setting and the search for new 'preclinical AD' biomarkers at even earlier disease stages, by using both mice and human samples. Ultimately, our findings may open new perspectives in identifying subjects at risk for AD significantly earlier, and in improving the stratification of patients for preventive treatment strategies.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Líquido Cefalorraquidiano/química , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Diagnóstico Precoce , Humanos , Estudos Longitudinais , Camundongos , Camundongos Transgênicos
13.
Brain Pathol ; 25(6): 743-52, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25677332

RESUMO

An important early event in the pathogenesis of Alzheimer's disease (AD) is the aberrant polymerization and extracellular accumulation of amyloid-ß peptide (Aß). In young transgenic mice expressing the human Aß-precursor protein (APP), deposits of Aß can be induced by the inoculation of minute amounts of brain extract containing Aß aggregates ("Aß seeds"), indicative of a prion-like seeding phenomenon. Moreover, focal intracerebral injection of Aß seeds can induce deposits not only in the immediate vicinity of the injection site, but, with time, also in distal regions of the brain. However, it remains uncertain whether the spatial progression of Aß deposits occurs via nonsystematic diffusion from the injection site to proximal regions or via directed transit along neuroanatomical pathways. To address this question, we analyzed the spatiotemporal emergence of Aß deposits in two different APP-transgenic mouse models that had been previously inoculated with Aß seeds into the hippocampal formation. The results revealed a specific, neuroanatomically constrained pattern of induced Aß deposits in structures corresponding to the limbic connectome, supporting the hypothesis that neuronal pathways act as conduits for the movement of proteopathic agents among brain regions, thereby facilitating the progression of disease.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Amiloidose/patologia , Conectoma , Sistema Límbico/metabolismo , Sistema Límbico/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Progressão da Doença , Humanos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação
14.
Science ; 330(6006): 980-2, 2010 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-20966215

RESUMO

The intracerebral injection of ß-amyloid-containing brain extracts can induce cerebral ß-amyloidosis and associated pathologies in susceptible hosts. We found that intraperitoneal inoculation with ß-amyloid-rich extracts induced ß-amyloidosis in the brains of ß-amyloid precursor protein transgenic mice after prolonged incubation times.


Assuntos
Peptídeos beta-Amiloides/química , Encéfalo/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/irrigação sanguínea , Química Encefálica , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Feminino , Injeções Intraperitoneais , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Príons/química , Príons/metabolismo , Dobramento de Proteína , Fatores de Tempo
15.
Ann Neurol ; 61(2): 153-61, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17212356

RESUMO

OBJECTIVE: Radiotherapy is an essential treatment modality for malignant gliomas, but it exerts adverse effects via promotion of glioma cell invasion in experimental glioma. Furthermore, irradiation induces vascular endothelial growth factor (VEGF) levels in gliomas, which is associated with poor prognosis. Here, we investigate the combination of the protein kinase C-beta inhibitor enzastaurin (ENZA) and radiotherapy in vitro and in vivo in comparison with either treatment alone. METHODS: We analyzed the effects of ENZA and irradiation on migration, apoptosis, and proliferation of glioma cells, as well as VEGF secretion in vitro. Neurotoxicity of ENZA was assessed in cerebellar granule neurons. After orthotopic intracerebral implantation of LNT-229 glioma cells in nude mice, the effects of in situ cerebral irradiation and oral application of ENZA on survival, tumor size, VEGF expression, apoptosis, and microvessel density in vivo were analyzed. RESULTS: Combining cerebral irradiation with ENZA leads to longer survival in vivo. ENZA diminishes tumor volume, irradiation-induced tumor satellite formation, upregulation of VEGF expression in vitro and in vivo, as well as enhanced microvessel density in vivo. Importantly, ENZA is not neurotoxic in vitro or in vivo. INTERPRETATION: Long-term administration of ENZA after radiotherapy is feasible and leads to long-term survival without neurotoxicity.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/radioterapia , Glioma/tratamento farmacológico , Glioma/radioterapia , Indóis/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/fisiopatologia , Terapia Combinada , Glioma/diagnóstico , Glioma/fisiopatologia , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Transplante de Neoplasias , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C beta , Análise de Sobrevida , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA