Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Virol ; 91(16)2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28592538

RESUMO

Intact and broad immune cell effector functions and specific individual cytokines have been linked to HIV disease outcome, but their relative contribution to HIV control remains unclear. We asked whether the proteome of secreted cytokines and signaling factors in peripheral blood can be used to discover specific pathways critical for host viral control. A custom glass-based microarray, able to measure >600 plasma proteins involved in cell-to-cell communication, was used to measure plasma protein profiles in 96 HIV-infected, treatment-naive individuals with high (>50,000) or low (<10,000 HIV RNA copies/ml) viral loads. Univariate and regression model analysis demonstrate that plasma levels of soluble interleukin-27 (IL-27) are significantly elevated in individuals with high plasma viremia (P < 0.0001) and are positively correlated with proviral HIV-DNA copy numbers in peripheral blood mononuclear cells (PBMC) (Rho = 0.4011; P = 0.0027). Moreover, soluble IL-27 plasma levels are negatively associated with the breadth and magnitude of the total virus-specific T-cell responses and directly with plasma levels of molecules involved in Wnt/ß-catenin signaling. In addition to IL-27, gene expression levels of the specific IL-27 receptor (IL27RA) in PBMC correlated directly with both plasma viral load (Rho = 0.3531; P = 0.0218) and the proviral copy number in the peripheral blood as an indirect measure of partial viral reservoir (Rho = 0.4580; P = 0.0030). These results were validated in unrelated cohorts of early infected subjects as well as subjects before and after initiation of antiretroviral treatment, and they identify IL-27 and its specific receptor as a critical immune axis for the antiviral immune response and as robust correlates of viral load and proviral reservoir size in PBMC.IMPORTANCE The detailed knowledge of immune mechanisms that contribute to HIV control is a prerequisite for the design of effective treatment strategies to achieve HIV cure. Cells communicate with each other by secreting signaling proteins, and the blood is a key conduit for transporting such factors. Investigating the communication factors promoting effective immune responses and having potentially antiviral functions against HIV using a novel focused omics approach ("communicome") has the potential to significantly improve our knowledge of effective host immunity and accelerate the HIV cure agenda. Including 140 subjects with variable viral loads and measuring the plasma levels of >600 soluble proteins, our data highlight the importance of Th17 cells and Wnt/ß-catenin signaling in HIV control and especially identify the IL-27/IL-27 receptor subunit alpha (IL-27RA) axis as a predictor of plasma viral load and proviral copy number in the peripheral blood. These data may provide important guidance to therapeutic approaches in the HIV cure agenda.


Assuntos
Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV/imunologia , Interleucinas/metabolismo , Receptores de Interleucina/metabolismo , Carga Viral , Proteínas Sanguíneas/análise , Perfilação da Expressão Gênica , Humanos , Leucócitos Mononucleares/imunologia , Análise Serial de Proteínas
2.
Proc Natl Acad Sci U S A ; 106(29): 12145-50, 2009 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-19581601

RESUMO

A number of distinct beta-amyloid (Abeta) variants or multimers have been implicated in Alzheimer's disease (AD), and antibodies recognizing such peptides are in clinical trials. Humans have natural Abeta-specific antibodies, but their diversity, abundance, and function in the general population remain largely unknown. Here, we demonstrate with peptide microarrays the presence of natural antibodies against known toxic Abeta and amyloidogenic non-Abeta species in plasma samples and cerebrospinal fluid of AD patients and healthy controls aged 21-89 years. Antibody reactivity was most prominent against oligomeric assemblies of Abeta and pyroglutamate or oxidized residues, and IgGs specific for oligomeric preparations of Abeta1-42 in particular declined with age and advancing AD. Most individuals showed unexpected antibody reactivities against peptides unique to autosomal dominant forms of dementia (mutant Abeta, ABri, ADan) and IgGs isolated from plasma of AD patients or healthy controls protected primary neurons from Abeta toxicity. Aged vervets showed similar patterns of plasma IgG antibodies against amyloid peptides, and after immunization with Abeta the monkeys developed high titers not only against Abeta peptides but also against ABri and ADan peptides. Our findings support the concept of conformation-specific, cross-reactive antibodies that may protect against amyloidogenic toxic peptides. If a therapeutic benefit of Abeta antibodies can be confirmed in AD patients, stimulating the production of such neuroprotective antibodies or passively administering them to the elderly population may provide a preventive measure toward AD.


Assuntos
Envelhecimento/imunologia , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/imunologia , Anticorpos/imunologia , Fármacos Neuroprotetores/imunologia , Peptídeos/imunologia , Envelhecimento/efeitos dos fármacos , Doença de Alzheimer/sangue , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/toxicidade , Animais , Anticorpos/sangue , Anticorpos/líquido cefalorraquidiano , Citoproteção/efeitos dos fármacos , Demência/complicações , Demência/imunologia , Progressão da Doença , Genes Dominantes , Imunização , Imunoglobulina G/sangue , Camundongos , Peso Molecular , Neurônios/citologia , Neurônios/efeitos dos fármacos , Peptídeos/química , Primatas/imunologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Quaternária de Proteína
3.
Neurochem Int ; 39(5-6): 393-400, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11578774

RESUMO

Recent studies have implicated pro- and anti-inflammatory cytokines as integral to Alzheimer's disease (AD) pathogenesis. Among them, transforming growth factor-beta (TGF-beta) is emerging as an important factor in regulating inflammatory responses. This multifunctional cytokine might be centrally involved in several aspects of AD pathogenesis by regulating beta-amyloid precursor protein synthesis and processing, plaque formation, astroglial and microglial response and neuronal cell death. Among all of these potential roles, studies in transgenic and infusion animal models have shown that TGF-beta may primarily contribute to AD pathogenesis by influencing A beta production and deposition, which in turn might result in damage to the brain microvasculature. The lessons learned from these models are of great interest not only for understanding of the role of TGF-beta in AD, but also for future treatments where testing of anti-inflammatory agents such as ibuprofen and an amyloid vaccine hold great promise. In this regard, further elucidation of the signal pathways by which TGF-beta exerts its effect in AD might lead to specific targets for further therapeutic intervention.


Assuntos
Doença de Alzheimer/patologia , Circulação Cerebrovascular , Fator de Crescimento Transformador beta/fisiologia , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Vasos Sanguíneos/patologia , Humanos , Camundongos , Camundongos Transgênicos
4.
Proc Natl Acad Sci U S A ; 98(15): 8838-43, 2001 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-11447277

RESUMO

Human apolipoprotein (apo) E4, a major risk factor for Alzheimer's disease (AD), occurs in amyloid plaques and neurofibrillary tangles (NFTs) in AD brains; however, its role in the pathogenesis of these lesions is unclear. Here we demonstrate that carboxyl-terminal-truncated forms of apoE, which occur in AD brains and cultured neurons, induce intracellular NFT-like inclusions in neurons. These cytosolic inclusions were composed of phosphorylated tau, phosphorylated neurofilaments of high molecular weight, and truncated apoE. Truncated apoE4, especially apoE4(Delta 272--299), induced inclusions in up to 75% of transfected neuronal cells, but not in transfected nonneuronal cells. ApoE4 was more susceptible to truncation than apoE3 and resulted in much greater intracellular inclusion formation. These results suggest that apoE4 preferentially undergoes intracellular processing, creating a bioactive fragment that interacts with cytoskeletal components and induces NFT-like inclusions containing phosphorylated tau and phosphorylated neurofilaments of high molecular weight in neurons.


Assuntos
Doença de Alzheimer/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Emaranhados Neurofibrilares/metabolismo , Fragmentos de Peptídeos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Apolipoproteína E4 , Apolipoproteínas E/fisiologia , Encéfalo/patologia , Células Cultivadas , Citosol/metabolismo , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Líquido Intracelular/metabolismo , Mutagênese , Emaranhados Neurofibrilares/patologia , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , Fragmentos de Peptídeos/fisiologia , Proteínas tau/metabolismo
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
8.
Neurobiol Aging ; 21(3): 383-421, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10858586

RESUMO

Inflammation clearly occurs in pathologically vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insoluble abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insoluble amyloid beta peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclinical to terminal stages of AD, local upregulation of complement, cytokines, acute phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to significantly exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clinical studies, although still in their infancy, strongly suggest that AD inflammation significantly contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.


Assuntos
Doença de Alzheimer/patologia , Inflamação/patologia , Encéfalo/patologia , Humanos
9.
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
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
11.
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
12.
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
13.
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
14.
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
15.
Neurosci Lett ; 254(2): 97-100, 1998 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-9779929

RESUMO

Human immunodeficiency virus (HIV)-1 can invade the brain and cause degeneration of the central nervous system, resulting in a host of cognitive and motor impairments. HIV-1 glycoprotein 120 (gp120), has been implicated in the neurodegenerative effects of HIV infection. Here, gp120's neurotoxic potential is demonstrated in both transgenic mice and cultured cells. We observed that gp120 causes an induction of matrix metalloproteinase (MMP)-2 activity and protein in transgenic mouse brains and in transfected C6 cells. We propose that induced MMP-2 may contribute to a neurodegenerative environment by degrading extracellular matrix (ECM) fibronectin and type IV collagen.


Assuntos
Encéfalo/metabolismo , Gelatinases/metabolismo , Proteína gp120 do Envelope de HIV/fisiologia , Metaloendopeptidases/metabolismo , Animais , Linhagem Celular Transformada , Indução Enzimática/fisiologia , Proteína gp120 do Envelope de HIV/genética , Metaloproteinase 2 da Matriz , Camundongos , Camundongos Transgênicos/genética , Neurotoxinas/metabolismo , Ratos , Transfecção
16.
Nat Med ; 4(4): 441-6, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9546790

RESUMO

The human CD4 molecule (hCD4) is expressed on T lymphocytes and macrophages and acts as a key component of the cellular receptor for HIV. At baseline, hCD4 transgenic mice expressed hCD4 on microglia, the resident mononuclear phagocytes of the brain, and showed no neuronal damage. Activation of brain microglia by peripheral immune challenges elicited neurodegeneration in hCD4 mice but not in nontransgenic controls. In post-mortem brain tissues from AIDS patients with opportunistic infections, but without typical HIV encephalitis, hCD4 expression correlated with neurodegeneration. We conclude that hCD4 may function as an important mediator of indirect neuronal damage in infectious and immune-mediated diseases of the central nervous system.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/imunologia , Infecções Oportunistas Relacionadas com a AIDS/patologia , Antígenos CD/fisiologia , Encéfalo/imunologia , Antígenos CD4/fisiologia , Microglia/imunologia , Degeneração Neural/imunologia , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Encéfalo/patologia , Antígenos CD4/biossíntese , Antígenos CD4/genética , Heterozigoto , Homozigoto , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Microglia/patologia , Neocórtex/imunologia , Neocórtex/patologia , Degeneração Neural/patologia , Sinapses/patologia
17.
Nature ; 389(6651): 603-6, 1997 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-9335500

RESUMO

Deposition of amyoid-beta peptide in the central nervous system is a hallmark of Alzheimer's disease and a possible cause of neurodegeneration. The factors that initiate or promote deposition of amyloid-beta peptide are not known. The transforming growth factor TGF-beta1 plays a central role in the response of the brain to injury, and increased TGF-beta1 has been found in the central nervous system of patients with Alzheimer's disease. Here we report that TGF-beta1 induces amyloid-beta deposition in cerebral blood vessels and meninges of aged transgenic mice overexpressing this cytokine from astrocytes. Co-expression of TGF-beta1 in transgenic mice overexpressing amyloid-precursor protein, which develop Alzheimer's like pathology, accelerated the deposition of amyloid-beta peptide. More TGF-beta1 messenger RNA was present in post-mortem brain tissue of Alzheimer's patients than in controls, the levels correlating strongly with amyloid-beta deposition in the damaged cerebral blood vessels of patients with cerebral amyloid angiopathy. These results indicate that overexpression of TGF-beta1 may initiate or promote amyloidogenesis in Alzheimer's disease and in experimental models and so may be a risk factor for developing Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Amiloidose/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Idoso , Envelhecimento/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Amiloidose/patologia , Animais , Astrócitos/metabolismo , Benzotiazóis , Encéfalo/metabolismo , Encéfalo/patologia , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Tiazóis/metabolismo
18.
J Neuroimmunol ; 77(1): 45-50, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9209267

RESUMO

Cerebral expression of the injury response cytokine transforming growth factor-beta 1 (TGF-beta 1) has been found to be increased in several neurological diseases but it remains unclear whether its function is primarily beneficial or detrimental. Here we show that transgenic (tg) mice that overexpress bioactive (TGF-beta 1 in the central nervous system (CNS) and show no overt phenotype in the unmanipulated state, are more susceptible to the immune-mediated CNS disease experimental autoimmune encephalomyelitis (EAE). TGF-beta 1 tg mice with EAE showed an earlier onset of clinical symptoms, more severe disease and increased mononuclear cell infiltration in their spinal cords compared with non-tg littermate controls with EAE. Whereas previous observations indicated that increased peripheral levels of TGF-beta 1 can suppress EAE, our findings demonstrate that local expression of TGF-beta 1 within the CNS parenchyma can enhance immune cell infiltration and intensify the CNS impairment resulting from peripherally triggered autoimmune responses.


Assuntos
Astrócitos/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Antígenos/farmacologia , Astrócitos/imunologia , Encéfalo/citologia , Encéfalo/imunologia , Encéfalo/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Mutagênese/imunologia , Medula Espinal/citologia , Medula Espinal/imunologia , Medula Espinal/patologia , Fator de Crescimento Transformador beta/genética
19.
Brain Res Brain Res Rev ; 23(1-2): 47-61, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9063586

RESUMO

beta-Amyloid precursor protein (beta APP), transforming growth factor beta (TGF beta), and tumor necrosis factor-alpha (TNF alpha) are remarkably pleiotropic neural cytokines/neurotrophic factors that orchestrate intricate injury-related cellular and molecular interactions. The links between these three factors include: their responses to injury; their interactive effects on astrocytes, microglia and neurons; their ability to induce cytoprotective responses in neurons; and their association with cytopathological alterations in Alzheimer's disease. Astrocytes and microglia each produce and respond to TGF beta and TNF alpha in characteristic ways when the brain is injured. TGF beta, TNF alpha and secreted forms of beta APP (sAPP) can protect neurons against excitotoxic, metabolic and oxidative insults and may thereby serve neuroprotective roles. On the other hand, under certain conditions TNF alpha and the fibrillogenic amyloid beta-peptide (A beta) derivative of beta APP can promote damage of neuronal and glial cells, and may play roles in neurodegenerative disorders. Studies of genetically manipulated mice in which TGF beta, TNF alpha or beta APP ligand or receptor levels are altered suggest important roles for each factor in cellular responses to brain injury and indicate that mediators of neural injury responses also have the potential to enhance amyloidogenesis and/or to interfere with neuroregeneration if expressed at abnormal levels or modified by strategic point mutations. Recent studies have elucidated signal transduction pathways of TGF beta (serine/threonine kinase cascades), TNF alpha (p55 receptor linked to a sphingomyelin-ceramide-NF kappa B pathway), and secreted forms of beta APP (sAPP; receptor guanylate cyclase-cGMP-cGMP-dependent kinase-K+ channel activation). Knowledge of these signaling pathways is revealing novel molecular targets on which to focus neuroprotective therapeutic strategies in disorders ranging from stroke to Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/fisiologia , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Lesões Encefálicas/patologia , Humanos
20.
J Clin Invest ; 97(3): 789-98, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8609236

RESUMO

HIV-1 associated central nervous system (CNS) disease involves neuronal damage and prominent reactive astrocytosis, the latter characterized by strong upregulation of the glial fibrillary acidic protein (GFAP) in astrocytes. Similar alterations are found in transgenic mice expressing the HIV-1 envelope protein gp120 in the CNS. Because alterations of astrocyte functions could contribute to neuronal impairment, we compared brains of gp120 transgenic mice and gp120-transfected C6 astrocytoma cells with controls and found that gp120 induced a prominent elevation of steady state GFAP mRNA levels, primarily due to transcript stabilization. Increased levels of GFAP mRNA were also found in nontransfected C6 cells exposed to recombinant gp120. Exposure of C6 cells or primary mouse astrocytes to soluble gp120 led to activation of PKC as indicated by redistribution and increase in PKC immunoreactivity at the single cell level. gp120 effects were diminished by inhibitors of protein kinase C (PKC) but not inhibitors of protein kinase A. PKC activity was upmodulated in gp120-transfected C6 cells and in the CNS of gp120 transgenic mice. Further, brain tissue from patients with HIV-1 encephalitis and from gp120 transgenic mice showed increased PKC immunoreactivity. Taken together, these results indicate that gp120-induced increases in PKC activity may contribute to the gliosis seen in gp120 transgenic mice as well as in HIV-1-infected humans and raise the question of whether dysregulation of signal transduction pathways represents a general mechanism of HIV-associated pathogenesis.


Assuntos
Complexo AIDS Demência/etiologia , Astrócitos/patologia , Proteína gp120 do Envelope de HIV/toxicidade , HIV-1/patogenicidade , Transdução de Sinais , Animais , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/biossíntese , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/isolamento & purificação , Proteína gp120 do Envelope de HIV/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...