Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Br J Haematol ; 196(3): 639-648, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34671975

RESUMO

Relapsed/refractory multiple myeloma (RRMM) is known to have a high burden of disease and complications associated with refractoriness to prior lines of therapy. Severe pain and fatigue symptoms and impairments in physical and emotional functioning have been strongly linked to reduced health-related quality of life (HRQoL) in patients with RRMM. Assessment of patient reported-outcome measures from the pivotal, Phase II HORIZON study (OP-106; NCT02963493) in patients with RRMM (n = 64) demonstrated that melphalan flufenamide (melflufen) plus dexamethasone treatment preserved HRQoL. Patients had clinically meaningful improvements, even after eight treatment cycles, in relevant scales such as global health status/QoL, physical functioning, emotional functioning, pain, and fatigue. Patients with triple-class-refractory disease (n = 50) displayed similar improvements. Patient-reported outcome deterioration was delayed for a substantial amount of time in patients who experienced a response to melflufen plus dexamethasone treatment relative to patients who did not experience a response. These findings support the notion that treatment with melflufen plus dexamethasone may sustain or improve HRQoL over time in patients with RRMM, including in patients with triple-class-refractory disease for whom outcomes are generally worse. The clinical benefits observed in patients from the HORIZON trial are encouraging and supportive of translation into real-world practice.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores Tumorais , Dexametasona/administração & dosagem , Feminino , Humanos , Masculino , Melfalan/administração & dosagem , Pessoa de Meia-Idade , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/epidemiologia , Mieloma Múltiplo/etiologia , Gradação de Tumores , Estadiamento de Neoplasias , Medidas de Resultados Relatados pelo Paciente , Resultado do Tratamento
2.
BMC Health Serv Res ; 22(1): 1020, 2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-35948977

RESUMO

BACKGROUND: Due to unwanted delays and suboptimal resource control of helicopter emergency medical services (HEMS), regional HEMS coordinators have recently been introduced in Norway. This may represent an unnecessary link in the alarm chain, which could cause delays in HEMS dispatch. Systematic evaluations of this intervention are lacking. We wanted to conduct this study to assess possible changes in HEMS response times, mission distribution patterns and patient characteristics within our region following this intervention. METHODS: We retrospectively collected timeline parameters, patient characteristics and GPS positions from HEMS missions executed by three regional HEMS bases in Mid-Norway during 2017-2018 (preintervention) and 2019 (postintervention). The mean regional response time in HEMS missions was assessed by an interrupted time series analysis (ITS). The geographical mission distribution between regional HEMS resources was assessed by a before-after study with a convex hull-based method. RESULTS: There was no significant change in the level (-0.13 min/month, p = 0.88) or slope (-0.13 min/month, p = 0.30) of the mean regional response time trend line pre- and postintervention. For one HEMS base, the service area was increased, and the median mission distance was significantly longer. For the two other bases, the service areas were reduced. Both the mean NACA score (4.13 ± SD 0.027 vs 3.98 ± SD 0.04, p < 0.01) and the proportion of patients with severe illness or injury (NACA 4-7, 68.2% vs 61.5%, p < 0.001) were higher in the postintervention group. CONCLUSION: The introduction of a regional HEMS coordinator in Mid-Norway did not cause prolonged response times in acute HEMS missions during the first year after implementation. Higher NACA scores in the patients treated postintervention suggest better selection of HEMS use.


Assuntos
Resgate Aéreo , Serviços Médicos de Emergência , Aeronaves , Serviços Médicos de Emergência/métodos , Humanos , Tempo de Reação , Estudos Retrospectivos
3.
Biochim Biophys Acta ; 1862(10): 1847-60, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27425031

RESUMO

Amyloid plaques in Alzheimer's disease (AD) mice are surrounded by activated microglia. The functional role of microglia activation in AD is not well understood; both detrimental and beneficial effects on AD progression have been reported. Here we show that the population of activated microglia in the cortex of the APPswe/PS1dE9 mouse AD model is divided into a CD11c-positive and a CD11c-negative subpopulation. Cd11c transcript levels and number of CD11c-positive microglia increase sharply when plaques start to occur and both parameters continue to rise in parallel with the age-related increasing plaque load. CD11c cells are localized near plaques at all stages of the disease development and constitute 23% of all activated microglia. No differences between these two populations were found in terms of proliferation, immunostaining intensity of Iba1, MHC class II, CD45, or immunoproteasome subunit LMP7/ß5i. Comparison of the transcriptome of isolated CD11c-positive and CD11c-negative microglia from the cortex of aged APPswe/PS1dE9 with WT microglia showed that gene expression changes had a similar general pattern. However, a differential expression was found for genes involved in immune signaling (Il6, S100a8/Mrp8, S100a9/Mrp14, Spp1, Igf1), lysosome activation, and carbohydrate- and cholesterol/lipid-metabolism (Apoe). In addition, the increased expression of Gpnmb/DC-HIL, Tm7sf4/DC-STAMP, and Gp49a/Lilrb4, suggests a suppressive/tolerizing influence of CD11c cells. We show that amyloid plaques in the APP/PS1 model are associated with two distinct populations of activated microglia: CD11c-positive and CD11c-negative cells. Our findings imply that CD11c-positive microglia can potentially counteract amyloid deposition via increased Aß-uptake and degradation, and by containing the inflammatory response.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Antígeno CD11c/metabolismo , Regulação da Expressão Gênica , Microglia/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Antígeno CD11c/genética , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Camundongos , Camundongos Transgênicos , Microglia/patologia , Proteínas do Tecido Nervoso/genética
4.
Glia ; 63(6): 1036-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25731615

RESUMO

Reactive astrocytes with an increased expression of intermediate filament (IF) proteins Glial Fibrillary Acidic Protein (GFAP) and Vimentin (VIM) surround amyloid plaques in Alzheimer's disease (AD). The functional consequences of this upregulation are unclear. To identify molecular pathways coupled to IF regulation in reactive astrocytes, and to study the interaction with microglia, we examined WT and APPswe/PS1dE9 (AD) mice lacking either GFAP, or both VIM and GFAP, and determined the transcriptome of cortical astrocytes and microglia from 15- to 18-month-old mice. Genes involved in lysosomal degradation (including several cathepsins) and in inflammatory response (including Cxcl5, Tlr6, Tnf, Il1b) exhibited a higher AD-induced increase when GFAP, or VIM and GFAP, were absent. The expression of Aqp4 and Gja1 displayed the same pattern. The downregulation of neuronal support genes in astrocytes from AD mice was absent in GFAP/VIM null mice. In contrast, the absence of IFs did not affect the transcriptional alterations induced by AD in microglia, nor was the cortical plaque load altered. Visualizing astrocyte morphology in GFAP-eGFP mice showed no clear structural differences in GFAP/VIM null mice, but did show diminished interaction of astrocyte processes with plaques. Microglial proliferation increased similarly in all AD groups. In conclusion, absence of GFAP, or both GFAP and VIM, alters AD-induced changes in gene expression profile of astrocytes, showing a compensation of the decrease of neuronal support genes and a trend for a slightly higher inflammatory expression profile. However, this has no consequences for the development of plaque load, microglial proliferation, or microglial activation.


Assuntos
Doença de Alzheimer/metabolismo , Astrócitos/metabolismo , Proteína Glial Fibrilar Ácida/deficiência , Microglia/metabolismo , Vimentina/deficiência , Idoso , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Astrócitos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células/fisiologia , Quimiocina CXCL5/metabolismo , Modelos Animais de Doenças , Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Vimentina/genética
5.
Brain ; 136(Pt 5): 1415-31, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23604491

RESUMO

The proteasome is the major protein degradation system within the cell, comprised of different proteolytic subunits; amyloid-ß is thought to impair its activity in Alzheimer's disease. Neuroinflammation is a prominent hallmark of Alzheimer's disease, which may implicate an activation of the immunoproteasome, a specific proteasome variant induced by immune signalling that holds slightly different proteolytic properties than the constitutive proteasome. Using a novel cell-permeable proteasome activity probe, we found that amyloid-ß enhances proteasome activity in glial and neuronal cultures. Additionally, using a subunit-specific proteasome activity assay we showed that in the cortex of the APPswePS1dE9 plaque pathology mouse model, immunoproteasome activities were strongly increased together with increased messenger RNA and protein expression in reactive glia surrounding plaques. Importantly, this elevated activity was confirmed in human post-mortem tissue from donors with Alzheimer's disease. These findings are in contrast with earlier studies, which reported impairment of proteasome activity in human Alzheimer's disease tissue and mouse models. Targeting the increased immunoproteasome activity with a specific inhibitor resulted in a decreased expression of inflammatory markers in ex vivo microglia. This may serve as a potential novel approach to modulate sustained neuroinflammation and glial dysfunction associated with Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Neuroglia/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/imunologia , Animais , Células Cultivadas , Ativação Enzimática/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Neuroglia/imunologia , Células Tumorais Cultivadas
6.
Glia ; 60(4): 615-29, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22262260

RESUMO

Plaque deposition in Alzheimer's disease (AD) is known to decrease proliferation in neurogenic niches in AD mouse models, but the effects on cell proliferation and differentiation in other brain areas have not been studied in detail. We analyzed cell proliferation in the cortex of wild type (WT) and APPswePS1dE9 transgenic (AD) mice at different ages. Mice were studied shortly after the last BrdU injection (BrdU[ST]). In AD mice, the number of proliferating cells increased fourfold, coinciding with plaque appearance and its associated reactive gliosis and activation of microglia. An increase in the number of BrdU[ST]-cells expressing markers for activated microglia is underlying the enhanced proliferation. Cortical reactive astrocytes did not become proliferative since BrdU[ST]-cells were negative for different astrocyte-specific markers. The number of Olig2-positive oligodendrocyte precursor cells was unchanged. Four weeks after the last BrdU application, the number of BrdU[LT]-cells with an activated microglia signature was still enhanced in AD mice. None of the newborn cells had differentiated into oligodendrocytes, astrocytes, or neurons. On the basis of these observations, we conclude that amyloid plaque deposition increases proliferation of microglia around plaques but does not affect the proliferation of cortical oligodendrocyte precursor cells. No evidence was found for damage-induced proliferation of reactive astrocytes or for a redirected neurogenesis from the subventricular zone. The proliferation of microglia contributes to the rapid accumulation of microglia around plaques and may play a role in limitating plaque expansion.


Assuntos
Doença de Alzheimer/patologia , Proliferação de Células , Córtex Cerebral/patologia , Regulação da Expressão Gênica/genética , Fatores Etários , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bromodesoxiuridina/metabolismo , Complexo CD3/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/genética , Modelos Animais de Doenças , Gliose/etiologia , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Presenilina-1/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Deleção de Sequência/genética
7.
Acta Neuropathol Commun ; 7(1): 60, 2019 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-31023360

RESUMO

Inter-individual differences in cortisol production by the hypothalamus-pituitary-adrenal (HPA) axis are thought to contribute to clinical and pathological heterogeneity of multiple sclerosis (MS). At the same time, accumulating evidence indicates that MS pathogenesis may originate in the normal-appearing white matter (NAWM). Therefore, we performed a genome-wide transcriptional analysis, by Agilent microarray, of post-mortem NAWM of 9 control subjects and 18 MS patients to investigate to what extent gene expression reflects disease heterogeneity and HPA-axis activity. Activity of the HPA axis was determined by cortisol levels in cerebrospinal fluid and by numbers of corticotropin-releasing neurons in the hypothalamus, while duration of MS and time to EDSS6 served as indicator of disease severity. Applying weighted gene co-expression network analysis led to the identification of a range of gene modules with highly similar co-expression patterns that strongly correlated with various indicators of HPA-axis activity and/or severity of MS. Interestingly, molecular profiles associated with relatively mild MS and high HPA-axis activity were characterized by increased expression of genes that actively regulate inflammation and by molecules involved in myelination, anti-oxidative mechanism, and neuroprotection. Additionally, group-wise comparisons of gene expression in white matter from control subjects and NAWM from (subpopulations of) MS patients uncovered disease-associated gene expression as well as strongly up- or downregulated genes in patients with relatively benign MS and/or high HPA-axis activity, with many differentially expressed genes being previously undescribed in the context of MS. Overall, the data suggest that HPA-axis activity strongly impacts on molecular mechanisms in NAWM of MS patients, but partly also independently of disease severity.


Assuntos
Encéfalo/metabolismo , Hidrocortisona/genética , Esclerose Múltipla Crônica Progressiva/genética , Transcriptoma , Substância Branca/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Perfilação da Expressão Gênica , Humanos , Hidrocortisona/metabolismo , Pessoa de Meia-Idade , Esclerose Múltipla Crônica Progressiva/metabolismo , Índice de Gravidade de Doença
8.
Acta Neuropathol Commun ; 3: 31, 2015 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-26001565

RESUMO

INTRODUCTION: Microglia are tissue macrophages of the central nervous system that monitor brain homeostasis and react upon neuronal damage and stress. Aging and neurodegeneration induce a hypersensitive, pro-inflammatory phenotype, referred to as primed microglia. To determine the gene expression signature of priming, the transcriptomes of microglia in aging, Alzheimer's disease (AD), and amyotrophic lateral sclerosis (ALS) mouse models were compared using Weighted Gene Co-expression Network Analysis (WGCNA). RESULTS: A highly consistent consensus transcriptional profile of up-regulated genes was identified, which prominently differed from the acute inflammatory gene network induced by lipopolysaccharide (LPS). Where the acute inflammatory network was significantly enriched for NF-κB signaling, the primed microglia profile contained key features related to phagosome, lysosome, antigen presentation, and AD signaling. In addition, specific signatures for aging, AD, and ALS were identified. CONCLUSION: Microglia priming induces a highly conserved transcriptional signature with aging- and disease-specific aspects.


Assuntos
Envelhecimento/genética , Inflamação/genética , Microglia/imunologia , Doenças Neurodegenerativas/genética , Transdução de Sinais/genética , Transcriptoma/genética , Envelhecimento/imunologia , Doença de Alzheimer/genética , Animais , Humanos , Camundongos , NF-kappa B/genética , Doenças Neurodegenerativas/imunologia , Regulação para Cima
9.
PLoS One ; 10(9): e0138107, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26418157

RESUMO

Gaucher disease is characterized by lysosomal accumulation of glucosylceramide due to deficient activity of lysosomal glucocerebrosidase (GBA). In cells, glucosylceramide is also degraded outside lysosomes by the enzyme glucosylceramidase 2 (GBA2) of which inherited deficiency is associated with ataxias. The interest in GBA and glucosylceramide metabolism in the brain has grown following the notion that mutations in the GBA gene impose a risk factor for motor disorders such as α-synucleinopathies. We earlier developed a ß-glucopyranosyl-configured cyclophellitol-epoxide type activity based probe (ABP) allowing in vivo and in vitro visualization of active molecules of GBA with high spatial resolution. Labeling occurs through covalent linkage of the ABP to the catalytic nucleophile residue in the enzyme pocket. Here, we describe a method to visualize active GBA molecules in rat brain slices using in vivo labeling. Brain areas related to motor control, like the basal ganglia and motor related structures in the brainstem, show a high content of active GBA. We also developed a ß-glucopyranosyl cyclophellitol-aziridine ABP allowing in situ labeling of GBA2. Labeled GBA2 in brain areas can be identified and quantified upon gel electrophoresis. The distribution of active GBA2 markedly differs from that of GBA, being highest in the cerebellar cortex. The histological findings with ABP labeling were confirmed by biochemical analysis of isolated brain areas. In conclusion, ABPs offer sensitive tools to visualize active GBA and to study the distribution of GBA2 in the brain and thus may find application to establish the role of these enzymes in neurodegenerative disease conditions such as α-synucleinopathies and cerebellar ataxia.


Assuntos
Encéfalo/enzimologia , Doença de Gaucher/genética , Glucosilceramidase/metabolismo , Glucosilceramidas/metabolismo , Doenças Neurodegenerativas/genética , Animais , Astrócitos/enzimologia , Astrócitos/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Ataxia Cerebelar/genética , Ataxia Cerebelar/patologia , Imunofluorescência , Corantes Fluorescentes/química , Doença de Gaucher/patologia , Glucosilceramidase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/enzimologia , Microglia/metabolismo , Microscopia Confocal , Doenças Neurodegenerativas/patologia , Células de Purkinje/metabolismo , Ratos , Ratos Wistar
10.
Neurobiol Aging ; 35(1): 1-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23954174

RESUMO

Astrocytes and microglia become reactive in many neurological disorders resulting in phenotypic and functional alterations. Both cell types might also display functional changes during normal aging. To identify gene signatures and changes in basal cellular functions of astrocytes and microglia in relation to aging, we isolated viable astrocytes and microglia from young adult and aged mouse cortices and determined their gene expression profile. Aged astrocytes, compared with young astrocytes, showed an increased inflammatory phenotype and increased 'zinc ion binding.' Young astrocytes showed higher expression of genes involved in 'neuronal differentiation' and hemoglobin synthesis. Astrocyte expression of genes involved in neuronal signaling remains high throughout age. Aged microglia had higher expression of genes involved in 'vesicle release,' 'zinc ion binding,' and genes within the tumor necrosis factor-ligand family and young microglia had increased transcript levels of C-C motif chemokines. These data provide a transcriptome database of cell-type enriched genes of astrocytes and microglia from adult mice and give insight into the differential gene signature of astrocytes and microglia in relation to normal aging.


Assuntos
Envelhecimento/genética , Astrócitos/fisiologia , Separação Celular/métodos , Córtex Cerebral/citologia , Microglia/fisiologia , Transdução de Sinais/genética , Transcriptoma/genética , Animais , Astrócitos/citologia , Astrócitos/patologia , Diferenciação Celular/genética , Células Cultivadas , Quimiocinas CC/genética , Hemoglobinas/biossíntese , Inflamação/genética , Camundongos , Microglia/citologia , Microglia/patologia , Vesículas Secretórias/genética , Transdução de Sinais/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/genética , Transcrição Gênica/genética , Zinco/metabolismo
11.
Neurobiol Aging ; 35(12): 2746-2760, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25002035

RESUMO

Reactive astrocytes and microglia are associated with amyloid plaques in Alzheimer's disease (AD). Yet, not much is known about the molecular alterations underlying this reactive phenotype. To get an insight into the molecular changes underlying AD induced astrocyte and microglia reactivity, we performed a transcriptional analysis on acutely isolated astrocytes and microglia from the cortex of aged controls and APPswe/PS1dE9 AD mice. As expected, both cell types acquired a proinflammatory phenotype, which confirms the validity of our approach. Interestingly, we observed that the immune alteration in astrocytes was relatively more pronounced than in microglia. Concurrently, our data reveal that astrocytes display a reduced expression of neuronal support genes and genes involved in neuronal communication. The microglia showed a reduced expression of phagocytosis and/or endocytosis genes. Co-expression analysis of a human AD expression data set and the astrocyte and microglia data sets revealed that the inflammatory changes in astrocytes were remarkably comparable in mouse and human AD, whereas the microglia changes showed less similarity. Based on these findings we argue that chronically proinflammatory astrocyte and microglia phenotypes, showing a reduction of genes involved in neuronal support and neuronal signaling, are likely to contribute to the neuronal dysfunction and cognitive decline in AD.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/patologia , Inflamação/genética , Inflamação/patologia , Microglia/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Animais , Astrócitos/imunologia , Astrócitos/fisiologia , Separação Celular , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/patologia , Cognição , Modelos Animais de Doenças , Endocitose/genética , Expressão Gênica , Humanos , Camundongos Transgênicos , Microglia/imunologia , Microglia/fisiologia , Fagocitose/genética , Transmissão Sináptica/genética
12.
PLoS One ; 7(8): e42823, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912745

RESUMO

Glial fibrillary acidic protein (GFAP) is the main astrocytic intermediate filament (IF). GFAP splice isoforms show differential expression patterns in the human brain. GFAPδ is preferentially expressed by neurogenic astrocytes in the subventricular zone (SVZ), whereas GFAP(+1) is found in a subset of astrocytes throughout the brain. In addition, the expression of these isoforms in human brain material of epilepsy, Alzheimer and glioma patients has been reported. Here, for the first time, we present a comprehensive study of GFAP isoform expression in both wild-type and Alzheimer Disease (AD) mouse models. In cortex, cerebellum, and striatum of wild-type mice, transcripts for Gfap-α, Gfap-ß, Gfap-γ, Gfap-δ, Gfap-κ, and a newly identified isoform Gfap-ζ, were detected. Their relative expression levels were similar in all regions studied. GFAPα showed a widespread expression whilst GFAPδ distribution was prominent in the SVZ, rostral migratory stream (RMS), neurogenic astrocytes of the subgranular zone (SGZ), and subpial astrocytes. In contrast to the human SVZ, we could not establish an unambiguous GFAPδ localization in proliferating cells of the mouse SVZ. In APPswePS1dE9 and 3xTgAD mice, plaque-associated reactive astrocytes had increased transcript levels of all detectable GFAP isoforms and low levels of a new GFAP isoform, Gfap-ΔEx7. Reactive astrocytes in AD mice showed enhanced GFAPα and GFAPδ immunolabeling, less frequently increased vimentin and nestin, but no GFAPκ or GFAP(+1) staining. In conclusion, GFAPδ protein is present in SVZ, RMS, and neurogenic astrocytes of the SGZ, but also outside neurogenic niches. Furthermore, differential GFAP isoform expression is not linked with aging or reactive gliosis. This evidence points to the conclusion that differential regulation of GFAP isoforms is not involved in the reorganization of the IF network in reactive gliosis or in neurogenesis in the mouse brain.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Envelhecimento/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/genética , Animais , Especificidade de Anticorpos , Encéfalo/patologia , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida , Proteínas de Filamentos Intermediários/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
PLoS One ; 6(7): e21855, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21765921

RESUMO

The study of the neural basis of emotional empathy has received a surge of interest in recent years but mostly employing human neuroimaging. A simpler animal model would pave the way for systematic single cell recordings and invasive manipulations of the brain regions implicated in empathy. Recent evidence has been put forward for the existence of empathy in rodents. In this study, we describe a potential model of empathy in female rats, in which we studied interactions between two rats: a witness observes a demonstrator experiencing a series of footshocks. By comparing the reaction of witnesses with or without previous footshock experience, we examine the role of prior experience as a modulator of empathy. We show that witnesses having previously experienced footshocks, but not naïve ones, display vicarious freezing behavior upon witnessing a cage-mate experiencing footshocks. Strikingly, the demonstrator's behavior was in turn modulated by the behavior of the witness: demonstrators froze more following footshocks if their witness froze more. Previous experiments have shown that rats emit ultrasonic vocalizations (USVs) when receiving footshocks. Thus, the role of USV in triggering vicarious freezing in our paradigm is examined. We found that experienced witness-demonstrator pairs emitted more USVs than naïve witness-demonstrator pairs, but the number of USVs was correlated with freezing in demonstrators, not in witnesses. Furthermore, playing back the USVs, recorded from witness-demonstrator pairs during the empathy test, did not induce vicarious freezing behavior in experienced witnesses. Thus, our findings confirm that vicarious freezing can be triggered in rats, and moreover it can be modulated by prior experience. Additionally, our result suggests that vicarious freezing is not triggered by USVs per se and it influences back onto the behavior of the demonstrator that had elicited the vicarious freezing in witnesses, introducing a paradigm to study empathy as a social loop.


Assuntos
Empatia/fisiologia , Congelamento , Modelos Psicológicos , Animais , Comportamento Animal/fisiologia , Medo , Feminino , Humanos , Ratos , Ratos Long-Evans , Comportamento Social , Espectrografia do Som
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA