Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Cell Death Dis ; 6: e1725, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25880092

RESUMO

Disrupting particular mitochondrial fission and fusion proteins leads to the death of specific neuronal populations; however, the normal functions of mitochondrial fission in neurons are poorly understood, especially in vivo, which limits the understanding of mitochondrial changes in disease. Altered activity of the central mitochondrial fission protein dynamin-related protein 1 (Drp1) may contribute to the pathophysiology of several neurologic diseases. To study Drp1 in a neuronal population affected by Alzheimer's disease (AD), stroke, and seizure disorders, we postnatally deleted Drp1 from CA1 and other forebrain neurons in mice (CamKII-Cre, Drp1lox/lox (Drp1cKO)). Although most CA1 neurons survived for more than 1 year, their synaptic transmission was impaired, and Drp1cKO mice had impaired memory. In Drp1cKO cell bodies, we observed marked mitochondrial swelling but no change in the number of mitochondria in individual synaptic terminals. Using ATP FRET sensors, we found that cultured neurons lacking Drp1 (Drp1KO) could not maintain normal levels of mitochondrial-derived ATP when energy consumption was increased by neural activity. These deficits occurred specifically at the nerve terminal, but not the cell body, and were sufficient to impair synaptic vesicle cycling. Although Drp1KO increased the distance between axonal mitochondria, mitochondrial-derived ATP still decreased similarly in Drp1KO boutons with and without mitochondria. This indicates that mitochondrial-derived ATP is rapidly dispersed in Drp1KO axons, and that the deficits in axonal bioenergetics and function are not caused by regional energy gradients. Instead, loss of Drp1 compromises the intrinsic bioenergetic function of axonal mitochondria, thus revealing a mechanism by which disrupting mitochondrial dynamics can cause dysfunction of axons.


Assuntos
Região CA1 Hipocampal/fisiologia , Dinaminas/fisiologia , Mitocôndrias/metabolismo , Neurônios/fisiologia , Animais , Axônios/fisiologia , Região CA1 Hipocampal/metabolismo , Dinaminas/deficiência , Dinaminas/genética , Dinaminas/metabolismo , Metabolismo Energético , Feminino , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Sinapses/fisiologia
2.
Genes Brain Behav ; 7(3): 344-54, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17908176

RESUMO

The fibroblast growth factor family of secreted signaling molecules is essential for patterning in the central nervous system. Fibroblast growth factor 17 (Fgf17) has been shown to contribute to regionalization of the rodent frontal cortex. To determine how Fgf17 signaling modulates behavior, both during development and in adulthood, we studied mice lacking one or two copies of the Fgf17 gene. Fgf17-deficient mice showed no abnormalities in overall physical growth, activity level, exploration, anxiety-like behaviors, motor co-ordination, motor learning, acoustic startle, prepulse inhibition, feeding, fear conditioning, aggression and olfactory exploration. However, they displayed striking deficits in several behaviors involving specific social interactions. Fgf17-deficient pups vocalized less than wild-type controls when separated from their mother and siblings. Elimination of Fgf17 also decreased the interaction of adult males with a novel ovariectomized female in a social recognition test and reduced the amount of time opposite-sex pairs spent engaged in prolonged, affiliative interactions during exploration of a novel environment. After social exploration of a novel environment, Fgf17-deficient mice showed less activation of the immediate-early gene Fos in the frontal cortex than wild-type controls. Our findings show that Fgf17 is required for several complex social behaviors and suggest that disturbances in Fgf17 signaling may contribute to neuropsychiatric diseases that affect such behaviors.


Assuntos
Fatores de Crescimento de Fibroblastos/deficiência , Transtornos Mentais/genética , Comportamento Social , Animais , Cruzamentos Genéticos , Feminino , Lobo Frontal/fisiologia , Lobo Frontal/fisiopatologia , Regulação da Expressão Gênica , Genes fos , Habituação Psicofisiológica , Aprendizagem/fisiologia , Masculino , Comportamento Materno , Camundongos , Camundongos Knockout , Reconhecimento Psicológico
3.
Neurology ; 69(10): 986-97, 2007 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-17785667

RESUMO

BACKGROUND: The mechanisms underlying navigation impairments in Alzheimer disease (AD) are unknown. We characterized navigation in AD and mild cognitive impairment (MCI) to test the hypothesis that navigation disability reflects selective impairments in spatial cognition and relates to atrophy of specific brain regions. METHODS: We compared 13 mild AD and 21 MCI patients with 24 controls on a route-learning task that engaged various spatial processes. Using structural MRI and optimized voxel-based morphometry, we also investigated the neural correlates of spatial abilities in a subset of subjects (10 AD, 12 MCI, 21 controls). RESULTS: AD and MCI patients recognized landmarks as effectively as controls, but could not find their locations on maps or recall the order in which they were encountered. Half of AD and one-quarter of MCI patients got lost on the route, compared with less than 10% of controls. Regardless of diagnosis, patients who got lost had lower right posterior hippocampal and parietal volumes than patients and controls who did not get lost. The ability to identify locations on a map correlated with right posterior hippocampal and parietal volumes, whereas order memory scores correlated with bilateral inferior frontal volumes. CONCLUSIONS: The navigation disability in Alzheimer disease and mild cognitive impairment (MCI) involves a selective impairment of spatial cognition and is associated with atrophy of the right-lateralized navigation network. Extensive spatial impairments in MCI suggest that navigation tests may provide early markers of cognitive and neural damage.


Assuntos
Doença de Alzheimer/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Cognição/fisiologia , Desempenho Psicomotor/fisiologia , Comportamento Espacial/fisiologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Mapeamento Encefálico/métodos , Transtornos Cognitivos/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Rede Nervosa/patologia , Rede Nervosa/fisiologia , Percepção Espacial/fisiologia
4.
Proc Natl Acad Sci U S A ; 98(21): 12245-50, 2001 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-11572944

RESUMO

Alzheimer's disease and Parkinson's disease are associated with the cerebral accumulation of beta-amyloid and alpha-synuclein, respectively. Some patients have clinical and pathological features of both diseases, raising the possibility of overlapping pathogenetic pathways. We generated transgenic (tg) mice with neuronal expression of human beta-amyloid peptides, alpha-synuclein, or both. The functional and morphological alterations in doubly tg mice resembled the Lewy-body variant of Alzheimer's disease. These mice had severe deficits in learning and memory, developed motor deficits before alpha-synuclein singly tg mice, and showed prominent age-dependent degeneration of cholinergic neurons and presynaptic terminals. They also had more alpha-synuclein-immunoreactive neuronal inclusions than alpha-synuclein singly tg mice. Ultrastructurally, some of these inclusions were fibrillar in doubly tg mice, whereas all inclusions were amorphous in alpha-synuclein singly tg mice. beta-Amyloid peptides promoted aggregation of alpha-synuclein in a cell-free system and intraneuronal accumulation of alpha-synuclein in cell culture. beta-Amyloid peptides may contribute to the development of Lewy-body diseases by promoting the aggregation of alpha-synuclein and exacerbating alpha-synuclein-dependent neuronal pathologies. Therefore, treatments that block the production or accumulation of beta-amyloid peptides could benefit a broader spectrum of disorders than previously anticipated.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Deficiências da Aprendizagem/metabolismo , Transtornos da Memória/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson/metabolismo , Fatores Etários , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Sinucleínas , alfa-Sinucleína
5.
Nat Med ; 7(5): 612-8, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11329064

RESUMO

Abnormal accumulation of the amyloid-beta peptide (Abeta) in the brain appears crucial to pathogenesis in all forms of Alzheimer disease (AD), but the underlying mechanisms in the sporadic forms of AD remain unknown. Transforming growth factor beta1 (TGF-beta1), a key regulator of the brain's responses to injury and inflammation, has been implicated in Abeta deposition in vivo. Here we demonstrate that a modest increase in astroglial TGF-beta1 production in aged transgenic mice expressing the human beta-amyloid precursor protein (hAPP) results in a three-fold reduction in the number of parenchymal amyloid plaques, a 50% reduction in the overall Abeta load in the hippocampus and neocortex, and a decrease in the number of dystrophic neurites. In mice expressing hAPP and TGF-beta1, Abeta accumulated substantially in cerebral blood vessels, but not in parenchymal plaques. In human cases of AD, Abeta immunoreactivity associated with parenchymal plaques was inversely correlated with Abeta in blood vessels and cortical TGF-beta1 mRNA levels. The reduction of parenchymal plaques in hAPP/TGF-beta1 mice was associated with a strong activation of microglia and an increase in inflammatory mediators. Recombinant TGF-beta1 stimulated Abeta clearance in microglial cell cultures. These results demonstrate that TGF-beta1 is an important modifier of amyloid deposition in vivo and indicate that TGF-beta1 might promote microglial processes that inhibit the accumulation of Abeta in the brain parenchyma.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Microglia/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Vasos Sanguíneos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Ensaio de Imunoadsorção Enzimática , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1
6.
Am J Pathol ; 157(6): 2003-10, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11106573

RESUMO

Proteases and their inhibitors play key roles in physiological and pathological processes. Cerebral amyloid plaques are a pathological hallmark of Alzheimer's disease (AD). They contain amyloid-ss (Ass) peptides in tight association with the serine protease inhibitor alpha(1)-antichymotrypsin.(1,2) However, it is unknown whether the increased expression of alpha(1)-antichymotrypsin found in AD brains counteracts or contributes to the disease. We used regulatory sequences of the glial fibrillary acidic protein gene(3) to express human alpha(1)-antichymotrypsin (hACT) in astrocytes of transgenic mice. These mice were crossed with transgenic mice that produce human amyloid protein precursors (hAPP) and Ass in neurons.(4,5) No amyloid plaques were found in transgenic mice expressing hACT alone, whereas hAPP transgenic mice and hAPP/hACT doubly transgenic mice developed typical AD-like amyloid plaques in the hippocampus and neocortex around 6 to 8 months of age. Co-expression of hAPP and hACT significantly increased the plaque burden at 7 to 8, 14, and 20 months. Both hAPP and hAPP/hACT mice showed significant decreases in synaptophysin-immunoreactive presynaptic terminals in the dentate gyrus, compared with nontransgenic littermates. Our results demonstrate that hACT acts as an amyloidogenic co-factor in vivo and suggest that the role of hACT in AD is pathogenic.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/farmacologia , Astrócitos/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Inibidores de Serina Proteinase/farmacologia , alfa 1-Antiquimotripsina/farmacologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos/genética , Placa Amiloide/patologia , Inibidores de Serina Proteinase/genética , Sinapses/efeitos dos fármacos , Transgenes/genética , alfa 1-Antiquimotripsina/genética
8.
Neuroscience ; 97(2): 207-10, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10799751

RESUMO

Apolipoprotein E fulfills fundamental functions in lipid transport and neural tissue repair after injury.(6,8) Its three most common isoforms (E2, E3, and E4) are critical determinants of diverse human diseases, including major cardiovascular and neurodegenerative disorders.(8,14) Apolipoprotein E4 is associated with an increased risk for Alzheimer's disease(3,5) and poor clinical outcome after head injury or stroke.(11,16) The precise role of apolipoprotein E4 in these conditions remains unknown. To characterize the effects of human apolipoprotein E isoforms in vivo, we analysed transgenic Apoe knockout mice that express apolipoprotein E3 or E4 or both in the brain. Hemizygous and homozygous apolipoprotein E3 mice were protected against age-related and excitotoxin-induced neurodegeneration, whereas apolipoprotein E4 mice were not. Apolipoprotein E3/E4 bigenic mice were as susceptible to neurodegeneration as apolipoprotein E4 singly-transgenic mice. At eight months of age neurodegeneration was more severe in homozygous than in hemizygous apolipoprotein E4 mice consistent with a dose effect. Thus, apolipoprotein E4 is not only less neuroprotective than apolipoprotein E3 but also acts as a dominant negative factor that interferes with the beneficial function of apolipoprotein E3. The inhibition of this apolipoprotein E4 activity may be critical for the prevention and treatment of neurodegeneration in APOE varepsilon4 carriers.


Assuntos
Apolipoproteínas E/genética , Encéfalo/metabolismo , Doenças Neurodegenerativas/genética , Doença de Alzheimer/genética , Animais , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/deficiência , Apolipoproteínas E/fisiologia , Apolipoproteínas E/toxicidade , Axônios/patologia , Encéfalo/patologia , Dendritos/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/análise , Doenças Neurodegenerativas/patologia , Fármacos Neuroprotetores , Terminações Pré-Sinápticas/patologia , Sinaptofisina/análise
9.
J Neurosci ; 20(11): 4050-8, 2000 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10818140

RESUMO

Amyloid plaques are a neuropathological hallmark of Alzheimer's disease (AD), but their relationship to neurodegeneration and dementia remains controversial. In contrast, there is a good correlation in AD between cognitive decline and loss of synaptophysin-immunoreactive (SYN-IR) presynaptic terminals in specific brain regions. We used expression-matched transgenic mouse lines to compare the effects of different human amyloid protein precursors (hAPP) and their products on plaque formation and SYN-IR presynaptic terminals. Four distinct minigenes were generated encoding wild-type hAPP or hAPP carrying mutations that alter the production of amyloidogenic Abeta peptides. The platelet-derived growth factor beta chain promoter was used to express these constructs in neurons. hAPP mutations associated with familial AD (FAD) increased cerebral Abeta(1-42) levels, whereas an experimental mutation of the beta-secretase cleavage site (671(M-->I)) eliminated production of human Abeta. High levels of Abeta(1-42) resulted in age-dependent formation of amyloid plaques in FAD-mutant hAPP mice but not in expression-matched wild-type hAPP mice. Yet, significant decreases in the density of SYN-IR presynaptic terminals were found in both groups of mice. Across mice from different transgenic lines, the density of SYN-IR presynaptic terminals correlated inversely with Abeta levels but not with hAPP levels or plaque load. We conclude that Abeta is synaptotoxic even in the absence of plaques and that high levels of Abeta(1-42) are insufficient to induce plaque formation in mice expressing wild-type hAPP. Our results support the emerging view that plaque-independent Abeta toxicity plays an important role in the development of synaptic deficits in AD and related conditions.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/genética , Fragmentos de Peptídeos/biossíntese , Placa Amiloide/genética , Placa Amiloide/metabolismo , Sinapses/genética , Sinapses/fisiologia , Envelhecimento/patologia , Doença de Alzheimer/genética , Sequência de Aminoácidos , Peptídeos beta-Amiloides/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Degeneração Neural/genética , Fragmentos de Peptídeos/genética , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Receptores Pré-Sinápticos/genética , Receptores Pré-Sinápticos/metabolismo
10.
Ann N Y Acad Sci ; 903: 317-23, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10818521

RESUMO

Alzheimer's disease (AD) is frequently associated with cerebrovascular changes, including perivascular astrocytosis, amyloid deposition, and microvascular degeneration, but it is not known whether these pathological changes contribute to functional deficits in AD. To characterize the temporal relationship between amyloid deposition, cerebrovascular abnormalities, and potential functional changes, we studied transgenic mice that express transforming growth factor-beta 1 (TGF-beta 1) at low levels in astrocytes. TGF-beta 1 induced a prominent perivascular astrocytosis, followed by the accumulation of basement membrane proteins in microvessels, thickening of capillary basement membranes, and later, around 6 months of age, deposition of amyloid in cerebral blood vessels. At 9 months of age, various AD-like degenerative alterations were observed in endothelial cells and pericytes. Associated with these morphological changes were changes in regional cerebral glucose utilization. Preliminary results showed that TGF-beta 1 mice had significantly decreased glucose utilization in the mammillary bodies, structures involved in mnemonic and learning processes. Glucose utilization tended to be decreased in several other brain regions as well; however, in the inferior colliculus, it was markedly higher in TGF-beta 1 mice than in controls. We conclude that chronic overproduction of TGF-beta 1 triggers a pathogenic cascade leading to AD-like cerebrovascular amyloidosis, microvascular degeneration, and local alterations in brain metabolic activity. Similar mechanisms may be involved in AD pathogenesis.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Angiopatia Amiloide Cerebral/patologia , Circulação Cerebrovascular , Microcirculação/patologia , Fator de Crescimento Transformador beta/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Membrana Basal/patologia , Encéfalo/metabolismo , Capilares/patologia , Lobo Frontal/irrigação sanguínea , Lobo Frontal/patologia , Gliose , Humanos , Camundongos , Camundongos Transgênicos , Fator de Crescimento Transformador beta/fisiologia
12.
Science ; 287(5456): 1265-9, 2000 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-10678833

RESUMO

To elucidate the role of the synaptic protein alpha-synuclein in neurodegenerative disorders, transgenic mice expressing wild-type human alpha-synuclein were generated. Neuronal expression of human alpha-synuclein resulted in progressive accumulation of alpha-synuclein-and ubiquitin-immunoreactive inclusions in neurons in the neocortex, hippocampus, and substantia nigra. Ultrastructural analysis revealed both electron-dense intranuclear deposits and cytoplasmic inclusions. These alterations were associated with loss of dopaminergic terminals in the basal ganglia and with motor impairments. These results suggest that accumulation of wild-type alpha-synuclein may play a causal role in Parkinson's disease and related conditions.


Assuntos
Encéfalo/metabolismo , Dopamina/fisiologia , Corpos de Inclusão/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/ultraestrutura , Humanos , Corpos de Inclusão/ultraestrutura , Corpos de Lewy/ultraestrutura , Doença por Corpos de Lewy/metabolismo , Doença por Corpos de Lewy/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Microscopia Eletrônica , Atividade Motora , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Doenças Neurodegenerativas/patologia , Neurônios/ultraestrutura , Substância Negra/metabolismo , Substância Negra/ultraestrutura , Sinucleínas , Tirosina 3-Mono-Oxigenase/imunologia , Tirosina 3-Mono-Oxigenase/metabolismo , Ubiquitinas/metabolismo , alfa-Sinucleína
13.
J Neurosci ; 20(5): 2064-71, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10684907

RESUMO

Several neurological diseases are frequently accompanied by dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis. The HPA axis regulates the secretion of glucocorticoids (GCs), which play important roles in diverse brain functions, including cognition, emotion, and feeding. Under physiological conditions, GCs are adaptive and beneficial; however, prolonged elevations in GC levels may contribute to neurodegeneration and brain dysfunction. In the current study, we demonstrate that apolipoprotein E (apoE) deficiency results in age-dependent dysregulation of the HPA axis through a mechanism affecting primarily the adrenal gland. Apoe(-/-) mice, which develop neurodegenerative alterations as they age, had an age-dependent increase in basal adrenal corticosterone content and abnormally increased plasma corticosterone levels after restraint stress, whereas their plasma and pituitary adrenocorticotropin levels were either unchanged or lower than those in controls. HPA axis dysregulation was associated with behavioral and metabolic alterations. When anxiety levels were assessed in the elevated plus maze, Apoe(-/-) mice showed more anxiety than wild-type controls. Apoe(-/-) mice also showed reduced activity in the open field. Finally, Apoe(-/-) mice showed age-dependent increases in food and water intake, stomach and body weights, and decreases in brown and white adipose tissues. These results support a key role for apoE in the tonic inhibition of steroidogenesis and HPA axis activity and have important implications for the behavioral analysis of Apoe(-/-) mice.


Assuntos
Apolipoproteínas E/genética , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipotálamo-Hipofisário/fisiopatologia , Sistema Hipófise-Suprarrenal/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Glândulas Suprarrenais/química , Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Hormônio Adrenocorticotrópico/sangue , Hormônio Adrenocorticotrópico/química , Hormônio Adrenocorticotrópico/farmacologia , Animais , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Corticosterona/sangue , Ingestão de Líquidos , Ingestão de Alimentos , Metabolismo Energético/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Estresse Fisiológico/metabolismo , Estresse Fisiológico/fisiopatologia
14.
Am J Pathol ; 156(1): 139-50, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10623661

RESUMO

Cerebrovascular amyloid deposition and microvascular degeneration are frequently associated with Alzheimer's disease (AD), but the etiology and pathogenetic role of these abnormalities are unknown. Recently, transforming growth factor-beta1 (TGF-beta1) was implicated in cerebrovascular amyloid formation in transgenic mice with astroglial overproduction of TGF-beta1 and in AD. We tested whether TGF-beta1 overproduction induces AD-like cerebrovascular degeneration and analyzed how cerebrovascular abnormalities develop over time in TGF-beta1-transgenic mice. In cerebral microvessels from 3- to 4-month-old TGF-beta1-transgenic mice, which display a prominent perivascular astrocytosis, levels of the basement membrane proteins perlecan and fibronectin were severalfold higher than in vessels from nontransgenic mice. Consistent with this increase, cortical capillary basement membranes of TGF-beta1 mice were significantly thickened. These changes preceded amyloid deposition, which began at around 6 months of age. In 9- and 18-month-old TGF-beta1 mice, various degenerative changes in microvascular cells of the brain were observed. Endothelial cells were thinner and displayed abnormal, microvilli-like protrusions as well as occasional condensation of chromatin, and pericytes occupied smaller areas in capillary profiles than in nontransgenic controls. Similar cerebrovascular abnormalities have been reported in AD. We conclude that chronic overproduction of TGF-beta1 triggers an accumulation of basement membrane proteins and results in AD-like cerebrovascular amyloidosis and microvascular degeneration. Closely related processes may induce cerebrovascular pathology in AD.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/metabolismo , Vasos Sanguíneos/patologia , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Animais , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Circulação Cerebrovascular , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos/genética , Microcirculação/efeitos dos fármacos , Microscopia Eletrônica , Fatores de Tempo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/farmacologia
15.
Eur J Neurosci ; 11(12): 4398-402, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10594667

RESUMO

Transgenic mice expressing HIV-1 coat glycoprotein gp120 in brain glial cells were previously shown to display AIDS dementia-like neuropathological changes and reduced hippocampal long-term potentiation. In this report, neuromotor and cognitive performance in 3- and 12-month-old gp120-expressing mice was compared with wildtype controls. Rotarod and cage activity measures showed no significant differences between transgenic animals and controls of either age. Open field activity was slightly altered in 12-month-old gp120 animals (reduced corner crossings and dwell in centre), but not in the 3-month-olds. Cognitive assessment using the Morris water maze showed unimpaired performance in 3-month-old mice during acquisition and (no-platform) probe trials. In 12-month-old gp120 animals, escape latency and swimming velocity during the acquisition trials were significantly reduced, but performance improved at roughly the same rate as in control animals. However, the probe trials revealed a highly significant reduction in spatial retention in transgenic mice of this age. This demonstration of age-dependent impairments in open field activity and spatial reference memory may relate to cognitive and neuromotor deficits seen in a proportion of HIV-1-infected individuals.


Assuntos
Complexo AIDS Demência/fisiopatologia , Envelhecimento/fisiologia , Transtornos Cognitivos/fisiopatologia , Proteína gp120 do Envelope de HIV/biossíntese , HIV-1/genética , Complexo AIDS Demência/diagnóstico , Complexo AIDS Demência/genética , Animais , Comportamento Animal , Sintomas Comportamentais/diagnóstico , Sintomas Comportamentais/fisiopatologia , Transtornos Cognitivos/diagnóstico , Feminino , Proteína gp120 do Envelope de HIV/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Atividade Motora/fisiologia , Neuroglia/metabolismo , Equilíbrio Postural/fisiologia , Transtornos Psicomotores/diagnóstico , Transtornos Psicomotores/fisiopatologia , Percepção Espacial/fisiologia
16.
Am J Pathol ; 155(5): 1741-7, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10550330

RESUMO

The class A scavenger receptor (SR) is expressed on reactive microglia surrounding cerebral amyloid plaques in Alzheimer's disease (AD). Interactions between the SR and amyloid beta peptides (Abeta) in microglial cultures elicit phagocytosis of Abeta aggregates and release of neurotoxins. To assess the role of the SR in amyloid clearance and Abeta-associated neurodegeneration in vivo, we used the platelet-derived growth factor promoter to express human amyloid protein precursors (hAPPs) in neurons of transgenic mice. With increasing age, hAPP mice develop AD-like amyloid plaques. We bred heterozygous hAPP (hAPP(+/-)) mice that were wild type for SR (SR(+/+)) with SR knockout (SR(-/-)) mice. Crosses among the resulting hAPP(+/-)SR(+/-) offspring yielded hAPP(+/-) and hAPP(-/-) littermates that were SR(+/+) or SR(-/-). These second-generation mice were analyzed at 6 and 12 months of age for extent of cerebral amyloid deposition and loss of synaptophysin-immunoreactive presynaptic terminals. hAPP(-/-)SR(-/-) mice showed no lack of SR expression, plaque formation, or synaptic degeneration, indicating that lack of SR expression does not result in significant accumulation of endogenous amyloidogenic or neurotoxic factors. In hAPP(+/-) mice, ablation of SR expression did not alter number, extent, distribution, or age-dependent accumulation of plaques; nor did it affect synaptic degeneration. Our results do not support a critical pathogenic role for microglial SR expression in neurodegenerative alterations associated with cerebral beta amyloidosis.


Assuntos
Precursor de Proteína beta-Amiloide/fisiologia , Proteínas de Membrana , Placa Amiloide/patologia , Receptores Imunológicos/fisiologia , Receptores de Lipoproteínas , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/fisiologia , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Receptores Depuradores , Receptores Depuradores Classe A , Receptores Depuradores Classe B
17.
Brain Res ; 835(1): 91-5, 1999 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-10448200

RESUMO

Ablation of tissue regions, specific genes, or specific cell types represent important means of studying function in the nervous system. Here we summarize recent experience using a strategy for the genetically-targeted and conditionally regulated ablation of astroglial cells in different parts of the nervous system. The strategy is based on the targeted expression of herpes simplex virus thymidine kinase to astroglial cells using the glial fibrillary acid protein promoter in transgenic mice, combined with treatment with the antiviral agent ganciclovir. Under different experimental conditions we find that transgene-expressing astroglial cells can be selectively ablated by ganciclovir in the enteric nervous system, or in the injured forebrain or sciatic nerve, providing models in which to study the functions of these cells.


Assuntos
Astrócitos/fisiologia , Sistema Nervoso Central/fisiologia , Sistema Nervoso Entérico/fisiologia , Camundongos Transgênicos/fisiologia , Sistema Nervoso Periférico/fisiologia , Animais , Marcação de Genes , Camundongos , Camundongos Transgênicos/genética
18.
Neuron ; 23(2): 297-308, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10399936

RESUMO

Reactive astrocytes adjacent to a forebrain stab injury were selectively ablated in adult mice expressing HSV-TK from the Gfap promoter by treatment with ganciclovir. Injured tissue that was depleted of GFAP-positive astrocytes exhibited (1) a prolonged 25-fold increase in infiltration of CD45-positive leukocytes, including ultrastructurally identified monocytes, macrophages, neutrophils, and lymphocytes, (2) failure of blood-brain barrier (BBB) repair, (3) substantial neuronal degeneration that could be attenuated by chronic glutamate receptor blockade, and (4) a pronounced increase in local neurite outgrowth. These findings show that genetic targeting can be used to ablate scar-forming astrocytes and demonstrate roles for astrocytes in regulating leukocyte trafficking, repairing the BBB, protecting neurons, and restricting nerve fiber growth after injury in the adult central nervous system.


Assuntos
Astrócitos/patologia , Lesões Encefálicas/patologia , Movimento Celular , Leucócitos/patologia , Degeneração Neural/patologia , Neuritos/patologia , Ferimentos Perfurantes/patologia , Animais , Astrócitos/metabolismo , Barreira Hematoencefálica , Contagem de Células , Feminino , Ganciclovir/farmacologia , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Hipocampo/patologia , Histocitoquímica , Leucócitos/metabolismo , Camundongos , Camundongos Transgênicos , Neuritos/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/biossíntese , Timidina Quinase/genética
19.
Proc Natl Acad Sci U S A ; 96(13): 7547-52, 1999 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-10377452

RESUMO

Amyloid precursor proteins (APPs) are expressed in multiple organs and cell types in diverse species. Their conservation across species and high abundance in brain and the association of various APP missense mutations with autosomal dominant forms of familial Alzheimer's disease (FAD) suggest important roles for APP in the central nervous system. However, the basic functions of APP in the central nervous system remain largely unknown. To assess potential effects of APP on neuronal death and survival, we transfected APP-deficient rat neuroblastoma cells (B103) with DNA constructs encoding wild-type or FAD-mutant human APP. Wild-type, but not FAD-mutant, APP effectively protected cells against apoptosis induced by ultraviolet irradiation, staurosporine, or p53. Wild-type APP also strongly inhibited p53 DNA-binding activity and p53-mediated gene transactivation, whereas FAD-mutant APP did not. We conclude that APP protects neuronal cells against apoptosis by controlling p53 activation at the post-translational level. Disruption of this function by mutations or alterations in APP processing could enhance neuronal vulnerability to secondary insults and contribute to neuronal degeneration.


Assuntos
Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Apoptose/genética , Mutação , Proteína Supressora de Tumor p53/genética , Animais , Humanos , Neuroblastoma/genética , Neuroblastoma/patologia , Ratos , Células Tumorais Cultivadas
20.
J Neurosci ; 19(12): 4867-80, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10366621

RESUMO

Apolipoprotein (apo) E isoforms are key determinants of susceptibility to Alzheimer's disease. The apoE4 isoform is the major known genetic risk factor for this disease and is also associated with poor outcome after acute head trauma or stroke. To test the hypothesis that apoE3, but not apoE4, protects against age-related and excitotoxin-induced neurodegeneration, we analyzed apoE knockout (Apoe-/-) mice expressing similar levels of human apoE3 or apoE4 in the brain under control of the neuron-specific enolase promoter. Neuronal apoE expression was widespread in the brains of these mice. Kainic acid-challenged wild-type or Apoe-/- mice had a significant loss of synaptophysin-positive presynaptic terminals and microtubule-associated protein 2-positive neuronal dendrites in the neocortex and hippocampus, and a disruption of neurofilament-positive axons in the hippocampus. Expression of apoE3, but not of apoE4, protected against this excitotoxin-induced neuronal damage. ApoE3, but not apoE4, also protected against the age-dependent neurodegeneration seen in Apoe-/- mice. These differences in the effects of apoE isoforms on neuronal integrity may relate to the increased risk of Alzheimer's disease and to the poor outcome after head trauma and stroke associated with apoE4 in humans.


Assuntos
Apolipoproteínas E/genética , Química Encefálica/genética , Degeneração Neural/genética , Envelhecimento/metabolismo , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/genética , Animais , Apolipoproteína E3 , Apolipoproteína E4 , Apolipoproteínas E/análise , Apolipoproteínas E/líquido cefalorraquidiano , Dendritos/química , Dendritos/metabolismo , Ensaio de Imunoadsorção Enzimática , Agonistas de Aminoácidos Excitatórios , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Humanos , Ácido Caínico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/induzido quimicamente , Neurotoxinas/metabolismo , Terminações Pré-Sinápticas/química , Terminações Pré-Sinápticas/metabolismo , RNA Mensageiro/análise , Sinaptofisina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA