Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 295(48): 16251-16266, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-32913125

RESUMO

Several plant-derived compounds have demonstrated efficacy in pre-clinical Alzheimer's disease (AD) rodent models. Each of these compounds share a gallic acid (GA) moiety, and initial assays on this isolated molecule indicated that it might be responsible for the therapeutic benefits observed. To test this hypothesis in a more physiologically relevant setting, we investigated the effect of GA in the mutant human amyloid ß-protein precursor/presenilin 1 (APP/PS1) transgenic AD mouse model. Beginning at 12 months, we orally administered GA (20 mg/kg) or vehicle once daily for 6 months to APP/PS1 mice that have accelerated Alzheimer-like pathology. At 18 months of age, GA therapy reversed impaired learning and memory as compared with vehicle, and did not alter behavior in nontransgenic littermates. GA-treated APP/PS1 mice had mitigated cerebral amyloidosis, including brain parenchymal and cerebral vascular ß-amyloid deposits, and decreased cerebral amyloid ß-proteins. Beneficial effects co-occurred with reduced amyloidogenic and elevated nonamyloidogenic APP processing. Furthermore, brain inflammation, gliosis, and oxidative stress were alleviated. We show that GA simultaneously elevates α- and reduces ß-secretase activity, inhibits neuroinflammation, and stabilizes brain oxidative stress in a pre-clinical mouse model of AD. We further demonstrate that GA increases abundance of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10, Adam10) proprotein convertase furin and activates ADAM10, directly inhibits ß-site APP cleaving enzyme 1 (BACE1, Bace1) activity but does not alter Adam10 or Bace1 transcription. Thus, our data reveal novel post-translational mechanisms for GA. We suggest further examination of GA supplementation in humans will shed light on the exciting therapeutic potential of this molecule.


Assuntos
Proteína ADAM10/metabolismo , Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Ácido Gálico/farmacologia , Proteínas de Membrana/metabolismo , Proteína ADAM10/genética , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Modelos Animais de Doenças , Furina/genética , Furina/metabolismo , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Presenilina-1/genética , Presenilina-1/metabolismo
2.
J Biol Chem ; 294(8): 2714-2731, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30563837

RESUMO

"Nutraceuticals" are well-tolerated natural dietary compounds with drug-like properties that make them attractive as Alzheimer's disease (AD) therapeutics. Combination therapy for AD has garnered attention following a recent National Institute on Aging mandate, but this approach has not yet been fully validated. In this report, we combined the two most promising nutraceuticals with complementary anti-amyloidogenic properties: the plant-derived phenolics (-)-epigallocatechin-3-gallate (EGCG, an α-secretase activator) and ferulic acid (FA, a ß-secretase modulator). We used transgenic mice expressing mutant human amyloid ß-protein precursor and presenilin 1 (APP/PS1) to model cerebral amyloidosis. At 12 months of age, we orally administered EGCG and/or FA (30 mg/kg each) or vehicle once daily for 3 months. At 15 months, combined EGCG-FA treatment reversed cognitive impairment in most tests of learning and memory, including novel object recognition and maze tasks. Moreover, EGCG- and FA-treated APP/PS1 mice exhibited amelioration of brain parenchymal and cerebral vascular ß-amyloid deposits and decreased abundance of amyloid ß-proteins compared with either EGCG or FA single treatment. Combined treatment elevated nonamyloidogenic soluble APP-α and α-secretase candidate and down-regulated amyloidogenic soluble APP-ß, ß-C-terminal APP fragment, and ß-secretase protein expression, providing evidence for a shift toward nonamyloidogenic APP processing. Additional beneficial co-treatment effects included amelioration of neuroinflammation, oxidative stress, and synaptotoxicity. Our findings offer preclinical evidence that combined treatment with EGCG and FA is a promising AD therapeutic approach.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/fisiologia , Catequina/análogos & derivados , Disfunção Cognitiva/tratamento farmacológico , Ácidos Cumáricos/farmacologia , Modelos Animais de Doenças , Presenilina-1/fisiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Antioxidantes/farmacologia , Comportamento Animal , Catequina/farmacologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Quimioterapia Combinada , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
3.
J Biol Chem ; 292(27): 11310-11325, 2017 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-28512130

RESUMO

To date, there is no effective Alzheimer's disease (AD)-modifying therapy. Nonetheless, combination therapy holds promise, and nutraceuticals (natural dietary compounds with therapeutic properties) and their synthetic derivatives are well-tolerated candidates. We tested whether combination therapy with octyl gallate (OG) and ferulic acid (FA) improves cognition and mitigates AD-like pathology in the presenilin-amyloid ß-protein precursor (PSAPP) transgenic mouse model of cerebral amyloidosis. One-year-old mice with established ß-amyloid plaques received daily doses of OG and FA alone or in combination for 3 months. PSAPP mice receiving combination therapy had statistically significant improved cognitive function versus OG or FA single treatment on some (but not all) measures. We also observed additional statistically significant reductions in brain parenchymal and cerebral vascular ß-amyloid deposits as well as brain amyloid ß-protein abundance in OG- plus FA-treated versus singly-treated PSAPP mice. These effects coincided with enhanced nonamyloidogenic amyloid ß-protein precursor (APP) cleavage, increased α-secretase activity, and ß-secretase inhibition. We detected elevated expression of nonamyloidogenic soluble APP-α and the α-secretase candidate, a disintegrin and metalloproteinase domain-containing protein 10. Correspondingly, amyloidogenic ß-carboxyl-terminal APP fragment and ß-site APP-cleaving enzyme 1 expression levels were reduced. In parallel, the ratio of ß- to α-carboxyl-terminal APP fragment was decreased. OG and FA combination therapy strikingly attenuated neuroinflammation, oxidative stress, and synaptotoxicity. Co-treatment afforded additional statistically significant benefits on some, but not all, of these outcome measures. Taken together, these data provide preclinical proof-of-concept for AD combination therapy.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Cognição/efeitos dos fármacos , Ácidos Cumáricos/farmacologia , Ácido Gálico/análogos & derivados , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Ácido Gálico/farmacologia , Humanos , Camundongos , Camundongos Transgênicos
4.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795421

RESUMO

CD8+ T cells are crucial components of immunity and play a vital role in recovery from West Nile virus (WNV) infection. Here, we identify a previously unrecognized function of interleukin-17A (IL-17A) in inducing cytotoxic-mediator gene expression and promoting CD8+ T cell cytotoxicity against WNV infection in mice. We find that IL-17A-deficient (Il17a-/-) mice are more susceptible to WNV infection and develop a higher viral burden than wild-type (WT) mice. Interestingly, the CD8+ T cells isolated from Il17a-/- mice are less cytotoxic and express lower levels of cytotoxic-mediator genes, which can be restored by supplying recombinant IL-17A in vitro and in vivo Importantly, treatment of WNV-infected mice with recombinant IL-17A, as late as day 6 postinfection, significantly reduces the viral burden and increases survival, suggesting a therapeutic potential for IL-17A. In conclusion, we report a novel function of IL-17A in promoting CD8+ T cell cytotoxicity, which may have broad implications in other microbial infections and cancers. IMPORTANCE: Interleukin-17A (IL-17A) and CD8+ T cells regulate diverse immune functions in microbial infections, malignancies, and autoimmune diseases. IL-17A is a proinflammatory cytokine produced by diverse cell types, while CD8+ T cells (known as cytotoxic T cells) are major cells that provide immunity against intracellular pathogens. Previous studies have demonstrated a crucial role of CD8+ T cells in recovery from West Nile virus (WNV) infection. However, the role of IL-17A during WNV infection remains unclear. Here, we demonstrate that IL-17A protects mice from lethal WNV infection by promoting CD8+ T cell-mediated clearance of WNV. In addition, treatment of WNV-infected mice with recombinant IL-17A reduces the viral burden and increases survival of mice, suggesting a potential therapeutic. This novel IL-17A-CD8+ T cell axis may also have broad implications for immunity to other microbial infections and cancers, where CD8+ T cell functions are crucial.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Interleucina-17/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Febre do Nilo Ocidental/tratamento farmacológico , Vírus do Nilo Ocidental/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/virologia , Feminino , Expressão Gênica , Humanos , Interleucina-17/genética , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/virologia , Cultura Primária de Células , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Resultado do Tratamento , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/mortalidade , Febre do Nilo Ocidental/virologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/crescimento & desenvolvimento
6.
CNS Neurol Disord Drug Targets ; 14(5): 600-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25921740

RESUMO

Medulloblastoma, a tumor of the cerebellum, is the most common pediatric central nervous system malignancy. These tumors are etiologically linked to mutations in the Sonic hedgehog (Shh) pathway, which signals through the primary, non-motile cilium. The growth of these aggressive tumors relies on self-renewal of tumor-propagating cells known as cancer stem cells (CSCs). Previous reports have implicated CD133-expressing cells as CSCs in brain tumors, while those expressing CD15 have been shown to propagate medulloblastoma. Here, we demonstrate that CD133+ and CD15+ cells are distinct medulloblastoma populations. CD15+ cells comprise approximately 0.5-1% of total human medulloblastoma cells, display CSC properties in culture and are detected in the Smoothened A1 transgenic mouse model of medulloblastoma. Additionally, we report on a medulloblastoma patient with enriched CD15+ cells in recurrent vs primary medulloblastoma. We also demonstrate that human medulloblastoma cells critically rely on establishment of primary cilia to drive Shh-mediated cell division. Primary cilia are found in external granule cells of human fetal cerebellum and in 12/14 medulloblastoma samples. Yet, CD15+ medulloblastoma cells lack primary cilia, suggesting that this CSC population signals independently of Shh. These results are important when considering the effects of current and prospective treatment modalities on medulloblastoma CSC populations.


Assuntos
Neoplasias Encefálicas/patologia , Cílios/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Meduloblastoma/patologia , Células-Tronco Neoplásicas/patologia , Animais , Antígenos CD/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias Encefálicas/classificação , Bromodesoxiuridina , Feto , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Antígeno Ki-67/metabolismo , Imageamento por Ressonância Magnética , Meduloblastoma/classificação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Nestina/metabolismo , Neuropeptídeos/genética , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Receptor Smoothened , Fatores de Tempo , Fatores de Transcrição/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(13): E1577-86, 2015 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-25770217

RESUMO

Physiological processes rely on the regulation of total mRNA levels in a cell. In diploid organisms, the transcriptional activation of one or both alleles of a gene may involve trans-allelic interactions that provide a tight spatial and temporal level of gene expression regulation. The mechanisms underlying such interactions still remain poorly understood. Here, we demonstrate that lipopolysaccharide stimulation of murine macrophages rapidly resulted in the actin-mediated and transient homologous spatial proximity of Tnfα alleles, which was necessary for the mono- to biallelic switch in gene expression. We identified two new complementary long noncoding RNAs transcribed from the TNFα locus and showed that their knockdown had opposite effects in Tnfα spatial proximity and allelic expression. Moreover, the observed spatial proximity of Tnfα alleles depended on pyruvate kinase muscle isoform 2 (PKM2) and T-helper-inducing POZ-Krüppel-like factor (ThPOK). This study suggests a role for lncRNAs in the regulation of somatic homologous spatial proximity and allelic expression control necessary for fine-tuning mammalian immune responses.


Assuntos
Linfotoxina-alfa/genética , Linfotoxina-beta/genética , RNA Longo não Codificante , Ativação Transcricional , Fator de Necrose Tumoral alfa/genética , Alelos , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hibridização in Situ Fluorescente , Lipopolissacarídeos/química , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hormônios Tireóideos/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
8.
Neuron ; 85(3): 534-48, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25619654

RESUMO

The impact of inflammation suppressor pathways on Alzheimer's disease (AD) evolution remains poorly understood. Human genetic evidence suggests involvement of the cardinal anti-inflammatory cytokine, interleukin-10 (IL10). We crossed the APP/PS1 mouse model of cerebral amyloidosis with a mouse deficient in Il10 (APP/PS1(+)Il10(-/-)). Quantitative in silico 3D modeling revealed activated Aß phagocytic microglia in APP/PS1(+)Il10(-/-) mice that restricted cerebral amyloidosis. Genome-wide RNA sequencing of APP/PS1(+)Il10(-/-) brains showed selective modulation of innate immune genes that drive neuroinflammation. Il10 deficiency preserved synaptic integrity and mitigated cognitive disturbance in APP/PS1 mice. In vitro knockdown of microglial Il10-Stat3 signaling endorsed Aß phagocytosis, while exogenous IL-10 had the converse effect. Il10 deficiency also partially overcame inhibition of microglial Aß uptake by human Apolipoprotein E. Finally, the IL-10 signaling pathway was abnormally elevated in AD patient brains. Our results suggest that "rebalancing" innate immunity by blocking the IL-10 anti-inflammatory response may be therapeutically relevant for AD.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Imunidade Inata/imunologia , Interleucina-10/deficiência , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
9.
Am J Pathol ; 184(11): 2855-61, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25174876

RESUMO

Activated microglia are associated with amyloid plaques in transgenic mouse models of cerebral amyloidosis and in human Alzheimer disease; yet, their implication in Alzheimer disease pathogenesis remains unclear. It has been suggested that microglia play dual roles depending on the context of activation, contributing negatively to disease pathogenesis by secreting proinflammatory innate cytokines or performing a beneficial role via phagocytosis of amyloid beta (Aß) deposits. Toll-like receptors, most of which signal through the adaptor protein myeloid differentiation factor 88 (MyD88), have been suggested as candidate Aß innate pattern recognition receptors. It was recently reported that MyD88 deficiency reduced brain amyloid pathology and microglial activation. To assess a putative role of MyD88 in cerebral amyloidosis and glial activation in APPswe/PS1ΔE9 (APP/PS1) mice, we crossed MyD88-deficient (MyD88(-/-)) mice with APP/PS1 mice, interbred first filial offspring, and studied APP/PS1 MyD88(+/+), APP/PS1 MyD88(+/-), and APP/PS1 MyD88(-/-) cohorts. Biochemical analysis of detergent-soluble and detergent-insoluble Aß1-40 or Aß1-42 in brain homogenates did not reveal significant between-group differences. Furthermore, no significant differences were observed on amyloid plaque load or soluble fibrillar Aß by quantitative immunohistochemical analysis. In addition, neither activated microglia nor astrocytes differed among the three groups. These data suggest that MyD88 signaling is dispensable for Aß-induced glial activation and does not significantly affect the nature or extent of cerebral ß-amyloidosis in APP/PS1 mice.


Assuntos
Angiopatia Amiloide Cerebral/genética , Córtex Cerebral/metabolismo , Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Córtex Cerebral/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microglia/patologia , Fator 88 de Diferenciação Mieloide/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo
10.
Proc Natl Acad Sci U S A ; 111(33): E3458-66, 2014 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-25082897

RESUMO

Cancer cell secretion of TGF-ß is a potent mechanism for immune evasion. However, little is known about how central nervous system tumors guard against immune eradication. We sought to determine the impact of T-cell TGF-ß signaling blockade on progression of medulloblastoma (MB), the most common pediatric brain tumor. Genetic abrogation of T-cell TGF-ß signaling mitigated tumor progression in the smoothened A1 (SmoA1) transgenic MB mouse. T regulatory cells were nearly abolished and antitumor immunity was mediated by CD8 cytotoxic T lymphocytes. To define the CD8 T-cell subpopulation responsible, primed CD8 T cells were adoptively transferred into tumor-bearing immunocompromised SmoA1 recipients. This led to generation of CD8(+)/killer cell lectin-like receptor G1 high (KLRG1(hi))/IL-7R(lo) short-lived effector cells that expressed granzyme B at the tumor. These results identify a cellular immune mechanism whereby TGF-ß signaling blockade licenses the T-cell repertoire to kill pediatric brain tumor cells.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Comportamento Animal , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL
11.
J Biol Chem ; 289(44): 30303-30317, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25157105

RESUMO

Amyloid precursor protein (APP) proteolysis is required for production of amyloid-ß (Aß) peptides that comprise ß-amyloid plaques in the brains of patients with Alzheimer disease (AD). Here, we tested whether the experimental agent methylene blue (MB), used for treatment of methemoglobinemia, might improve AD-like pathology and behavioral deficits. We orally administered MB to the aged transgenic PSAPP mouse model of cerebral amyloidosis and evaluated cognitive function and cerebral amyloid pathology. Beginning at 15 months of age, animals were gavaged with MB (3 mg/kg) or vehicle once daily for 3 months. MB treatment significantly prevented transgene-associated behavioral impairment, including hyperactivity, decreased object recognition, and defective spatial working and reference memory, but it did not alter nontransgenic mouse behavior. Moreover, brain parenchymal and cerebral vascular ß-amyloid deposits as well as levels of various Aß species, including oligomers, were mitigated in MB-treated PSAPP mice. These effects occurred with inhibition of amyloidogenic APP proteolysis. Specifically, ß-carboxyl-terminal APP fragment and ß-site APP cleaving enzyme 1 protein expression and activity were attenuated. Additionally, treatment of Chinese hamster ovary cells overexpressing human wild-type APP with MB significantly decreased Aß production and amyloidogenic APP proteolysis. These results underscore the potential for oral MB treatment against AD-related cerebral amyloidosis by modulating the amyloidogenic pathway.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Amiloidose/tratamento farmacológico , Encefalopatias/tratamento farmacológico , Cognição/efeitos dos fármacos , Azul de Metileno/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/patologia , Amiloidose/psicologia , Animais , Encefalopatias/patologia , Encefalopatias/psicologia , Células CHO , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Cricetinae , Cricetulus , Avaliação Pré-Clínica de Medicamentos , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteólise
12.
J Neurosci ; 33(15): 6245-56, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575824

RESUMO

Alzheimer's disease (AD) is hallmarked by amyloid plaques, neurofibrillary tangles, and widespread cortical neuronal loss (Selkoe, 2001). The "amyloid cascade hypothesis" posits that cerebral amyloid sets neurotoxic events into motion that precipitate Alzheimer dementia (Hardy and Allsop, 1991). Yet, faithful recapitulation of all AD features in widely used transgenic (Tg) mice engineered to overproduce Aß peptides has been elusive. We have developed a Tg rat model (line TgF344-AD) expressing mutant human amyloid precursor protein (APPsw) and presenilin 1 (PS1ΔE9) genes, each independent causes of early-onset familial AD. TgF344-AD rats manifest age-dependent cerebral amyloidosis that precedes tauopathy, gliosis, apoptotic loss of neurons in the cerebral cortex and hippocampus, and cognitive disturbance. These results demonstrate progressive neurodegeneration of the Alzheimer type in these animals. The TgF344-AD rat fills a critical need for a next-generation animal model to enable basic and translational AD research.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/patologia , Transtornos Cognitivos/patologia , Hipocampo/patologia , Degeneração Neural/patologia , Placa Amiloide/patologia , Tauopatias/patologia , Fatores Etários , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal , Angiopatia Amiloide Cerebral , Córtex Cerebral/metabolismo , Transtornos Cognitivos/complicações , Transtornos Cognitivos/genética , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Feminino , Gliose/genética , Gliose/patologia , Hipocampo/metabolismo , Humanos , Masculino , Degeneração Neural/genética , Degeneração Neural/metabolismo , Placa Amiloide/genética , Presenilina-1/genética , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos , Tauopatias/metabolismo , Proteínas tau/metabolismo
13.
Biomed Res Int ; 2013: 740892, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23509770

RESUMO

Hematopoietic stem cells (HSCs) are unique in their capacity to give rise to all mature cells of the immune system. For years, HSC transplantation has been used for treatment of genetic and neoplastic diseases of the hematopoietic and immune systems. The sourcing of HSCs from human umbilical cord blood has salient advantages over isolation from mobilized peripheral blood. However, poor sample yield has prompted development of methodologies to expand HSCs ex vivo. Cytokines, trophic factors, and small molecules have been variously used to promote survival and proliferation of HSCs in culture, whilst strategies to lower the concentration of inhibitors in the culture media have recently been applied to promote HSC expansion. In this paper, we outline strategies to expand HSCs in vitro, and to improve engraftment and reconstitution of human immune systems in immunocompromised mice. To the extent that these "humanized" mice are representative of the endogenous human immune system, they will be invaluable tools for both basic science and translational medicine.


Assuntos
Técnicas de Cultura de Células/métodos , Modelos Animais de Doenças , Células-Tronco Hematopoéticas/citologia , Sistema Imunitário/fisiologia , Animais , Proliferação de Células , Sobrevivência Celular , Sangue Fetal/citologia , Antígenos HLA/metabolismo , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Transgênicos , Pesquisa Translacional Biomédica
14.
PLoS One ; 8(2): e55774, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23409038

RESUMO

Amyloid precursor protein (APP) proteolysis is required for production of amyloid-ß (Aß) peptides that comprise ß-amyloid plaques in brains of Alzheimer's disease (AD) patients. Recent AD therapeutic interest has been directed toward a group of anti-amyloidogenic compounds extracted from plants. We orally administered the brain penetrant, small molecule phenolic compound ferulic acid (FA) to the transgenic PSAPP mouse model of cerebral amyloidosis (bearing mutant human APP and presenilin-1 transgenes) and evaluated behavioral impairment and AD-like pathology. Oral FA treatment for 6 months reversed transgene-associated behavioral deficits including defective: hyperactivity, object recognition, and spatial working and reference memory, but did not alter wild-type mouse behavior. Furthermore, brain parenchymal and cerebral vascular ß-amyloid deposits as well as abundance of various Aß species including oligomers were decreased in FA-treated PSAPP mice. These effects occurred with decreased cleavage of the ß-carboxyl-terminal APP fragment, reduced ß-site APP cleaving enzyme 1 protein stability and activity, attenuated neuroinflammation, and stabilized oxidative stress. As in vitro validation, we treated well-characterized mutant human APP-overexpressing murine neuron-like cells with FA and found significantly decreased Aß production and reduced amyloidogenic APP proteolysis. Collectively, these results highlight that FA is a ß-secretase modulator with therapeutic potential against AD.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Comportamento Animal/efeitos dos fármacos , Ácidos Cumáricos/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Angiopatia Amiloide Cerebral , Ácidos Cumáricos/administração & dosagem , Ácidos Cumáricos/química , Modelos Animais de Doenças , Feminino , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo
15.
Mol Neurobiol ; 45(3): 564-70, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22644387

RESUMO

Although the discovery of cilia is one of the earliest in cell biology, the past two decades have witnessed an explosion of new insight into these enigmatic organelles. While long believed to be vestigial, cilia have recently moved into the spotlight as key players in multiple cellular processes, including brain development and homeostasis. This review focuses on the rapidly expanding basic biology of neural cilia, with special emphasis on the newly emerging B9 family of proteins. In particular, recent findings have identified a critical role for the B9 complex in a network of protein interactions that take place at the ciliary transition zone (TZ). We describe the essential role of these protein complexes in signaling cascades that require primary (nonmotile) cilia, including the sonic hedgehog pathway. Loss or dysfunction of ciliary trafficking and TZ function are linked to a number of neurologic diseases, which we propose to classify as neural ciliopathies. When taken together, the studies reviewed herein point to critical roles played by neural cilia, both in normal physiology and in disease.


Assuntos
Cílios/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Organogênese , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Cílios/patologia , Humanos , Sistema Nervoso/crescimento & desenvolvimento , Sistema Nervoso/metabolismo , Sistema Nervoso/patologia , Neurônios/patologia
16.
Immunity ; 36(3): 401-14, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22342844

RESUMO

We report that in the presence of signal 1 (NF-κB), the NLRP3 inflammasome was activated by mitochondrial apoptotic signaling that licensed production of interleukin-1ß (IL-1ß). NLRP3 secondary signal activators such as ATP induced mitochondrial dysfunction and apoptosis, resulting in release of oxidized mitochondrial DNA (mtDNA) into the cytosol, where it bound to and activated the NLRP3 inflammasome. The antiapoptotic protein Bcl-2 inversely regulated mitochondrial dysfunction and NLRP3 inflammasome activation. Mitochondrial DNA directly induced NLRP3 inflammasome activation, because macrophages lacking mtDNA had severely attenuated IL-1ß production, yet still underwent apoptosis. Both binding of oxidized mtDNA to the NLRP3 inflammasome and IL-1ß secretion could be competitively inhibited by the oxidized nucleoside 8-OH-dG. Thus, our data reveal that oxidized mtDNA released during programmed cell death causes activation of the NLRP3 inflammasome. These results provide a missing link between apoptosis and inflammasome activation, via binding of cytosolic oxidized mtDNA to the NLRP3 inflammasome.


Assuntos
Apoptose/imunologia , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , DNA Mitocondrial/imunologia , DNA Mitocondrial/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Animais , Expressão Gênica , Interleucina-1beta/biossíntese , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Oxirredução , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/imunologia , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Transdução de Sinais
17.
J Biol Chem ; 287(9): 6912-27, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22219198

RESUMO

Amyloid precursor protein (APP) proteolysis is essential for production of amyloid-ß (Aß) peptides that form ß-amyloid plaques in brains of Alzheimer disease (AD) patients. Recent focus has been directed toward a group of naturally occurring anti-amyloidogenic polyphenols known as flavonoids. We orally administered the flavonoid tannic acid (TA) to the transgenic PSAPP mouse model of cerebral amyloidosis (bearing mutant human APP and presenilin-1 transgenes) and evaluated cognitive function and AD-like pathology. Consumption of TA for 6 months prevented transgene-associated behavioral impairment including hyperactivity, decreased object recognition, and defective spatial reference memory, but did not alter nontransgenic mouse behavior. Accordingly, brain parenchymal and cerebral vascular ß-amyloid deposits and abundance of various Aß species including oligomers were mitigated in TA-treated PSAPP mice. These effects occurred with decreased cleavage of the ß-carboxyl-terminal APP fragment, lowered soluble APP-ß production, reduced ß-site APP cleaving enzyme 1 protein stability and activity, and attenuated neuroinflammation. As in vitro validation, we treated well characterized mutant human APP-overexpressing murine neuron-like cells with TA and found significantly reduced Aß production associated with less amyloidogenic APP proteolysis. Taken together, these results raise the possibility that dietary supplementation with TA may be prophylactic for AD by inhibiting ß-secretase activity and neuroinflammation and thereby mitigating AD pathology.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/prevenção & controle , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/prevenção & controle , Taninos/farmacologia , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular , Angiopatia Amiloide Cerebral/tratamento farmacológico , Angiopatia Amiloide Cerebral/patologia , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Encefalite/tratamento farmacológico , Encefalite/patologia , Feminino , Gliose/tratamento farmacológico , Gliose/patologia , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo
18.
J Virol ; 85(9): 4184-97, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21307196

RESUMO

Herpes simplex virus (HSV) infection is a classic example of latent viral infection in humans and experimental animal models. The HSV-1 latency-associated transcript (LAT) plays a major role in the HSV-1 latency reactivation cycle and thus in recurrent disease. Whether the presence of LAT leads to generation of dysfunctional T cell responses in the trigeminal ganglia (TG) of latently infected mice is not known. To address this issue, we used LAT-positive [LAT(+)] and LAT-deficient [LAT(-)] viruses to evaluate the effect of LAT on CD8 T cell exhaustion in TG of latently infected mice. The amount of latency as determined by quantitative reverse transcription-PCR (qRT-PCR) of viral DNA in total TG extracts was 3-fold higher with LAT(+) than with LAT(-) virus. LAT expression and increased latency correlated with increased mRNA levels of CD8, PD-1, and Tim-3. PD-1 is both a marker for exhaustion and a primary factor leading to exhaustion, and Tim-3 can also contribute to exhaustion. These results suggested that LAT(+) TG contain both more CD8(+) T cells and more CD8(+) T cells expressing the exhaustion markers PD-1 and Tim-3. This was confirmed by flow cytometry analyses of expression of CD3/CD8/PD-1/Tim-3, HSV-1, CD8(+) T cell pentamer (specific for a peptide derived from residues 498 to 505 of glycoprotein B [gB(498-505)]), interleukin-2 (IL-2), and tumor necrosis factor alpha (TNF-α). The functional significance of PD-1 and its ligands in HSV-1 latency was demonstrated by the significantly reduced amount of HSV-1 latency in PD-1- and PD-L1-deficient mice. Together, these results may suggest that both PD-1 and Tim-3 are mediators of CD8(+) T cell exhaustion and latency in HSV-1 infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , MicroRNAs/metabolismo , Gânglio Trigeminal/imunologia , Latência Viral , Animais , Antígenos de Diferenciação/biossíntese , Antígenos CD8/biossíntese , Citometria de Fluxo , Perfilação da Expressão Gênica , Receptor Celular 2 do Vírus da Hepatite A , Herpes Simples/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1 , Receptores Virais/biossíntese
19.
J Neurosci ; 31(4): 1355-65, 2011 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-21273420

RESUMO

Converging lines of evidence indicate dysregulation of the key immunoregulatory molecule CD45 (also known as leukocyte common antigen) in Alzheimer's disease (AD). We report that transgenic mice overproducing amyloid-ß peptide (Aß) but deficient in CD45 (PSAPP/CD45(-/-) mice) faithfully recapitulate AD neuropathology. Specifically, we find increased abundance of cerebral intracellular and extracellular soluble oligomeric and insoluble Aß, decreased plasma soluble Aß, increased abundance of microglial neurotoxic cytokines tumor necrosis factor-α and interleukin-1ß, and neuronal loss in PSAPP/CD45(-/-) mice compared with CD45-sufficient PSAPP littermates (bearing mutant human amyloid precursor protein and mutant human presenilin-1 transgenes). After CD45 ablation, in vitro and in vivo studies demonstrate an anti-Aß phagocytic but proinflammatory microglial phenotype. This form of microglial activation occurs with elevated Aß oligomers and neural injury and loss as determined by decreased ratio of anti-apoptotic Bcl-xL to proapoptotic Bax, increased activated caspase-3, mitochondrial dysfunction, and loss of cortical neurons in PSAPP/CD45(-/-) mice. These data show that deficiency in CD45 activity leads to brain accumulation of neurotoxic Aß oligomers and validate CD45-mediated microglial clearance of oligomeric Aß as a novel AD therapeutic target.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Antígenos Comuns de Leucócito/genética , Neurônios/patologia , Fragmentos de Peptídeos/metabolismo , Envelhecimento/metabolismo , Envelhecimento/patologia , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Barreira Hematoencefálica/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Humanos , Inflamação/patologia , Interleucina-1beta/metabolismo , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Microglia/imunologia , Microglia/patologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Presenilina-1/genética , Multimerização Proteica , Transgenes , Fator de Necrose Tumoral alfa/metabolismo
20.
Invest Ophthalmol Vis Sci ; 52(5): 2321-33, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21220560

RESUMO

PURPOSE: CD4(+)CD25(+)FoxP3(+) naturally occurring regulatory T cells (Tregs) maintain self-tolerance and function to suppress overly exuberant immune responses. However, it is unclear whether innate immune cells modulate Treg function. Here the authors examined the role of innate immunity in lymphomyeloid homeostasis. METHODS: The involvement of B cells, dendritic cells (DCs), macrophages, natural killer (NK) cells, and T cells in central nervous system (CNS) demyelination in different strains of mice infected ocularly with herpes simplex virus type 1 (HSV-1) was investigated. RESULTS: The authors found that depletion of macrophages, but not DCs, B cells, NK cells, CD4(+) T cells, or CD8(+) T cells, induced CNS demyelination irrespective of virus or mouse strain. As with macrophage depletion, mice deficient in interleukin (IL)-12p35 or IL-12p40 showed CNS demyelination after HSV-1 infection, whereas demyelination was undetectable in HSV-1-infected, IL-23p19-deficient, or Epstein-Barr virus-induced gene 3-deficient mice. Demyelination could be rescued in macrophage-depleted mice after the injection of IL-12p70 DNA and in IL-12p35(-/-) or IL-12p40(-/-) mice after injection with IL-12p35 or IL-12p40 DNA or with recombinant viruses expressing IL-12p35 or IL-12p40. Using FoxP3-, CD4-, CD8-, or CD25-depletion and gene-deficient mouse approaches, the authors demonstrated that HSV-1-induced demyelination was blocked in the absence of CD4, CD25, or FoxP3 in macrophage-depleted mice. Flow cytometry showed an elevation of CD4(+)CD25(+)FoxP3(+) T cells in the spleens of infected macrophage-depleted mice, and adoptive transfer of CD4(+)CD25(+) T cells to infected macrophage-depleted severe combined immunodeficient mice induced CNS demyelination. CONCLUSIONS: The authors demonstrated that macrophage IL-12p70 signaling plays an important role in maintaining immune homeostasis in the CNS by preventing the development of autoaggressive CD4(+) Tregs.


Assuntos
Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/prevenção & controle , Herpesvirus Humano 1/patogenicidade , Interleucina-12/metabolismo , Macrófagos/fisiologia , Transdução de Sinais/fisiologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Autoimunidade/imunologia , Linfócitos B/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/virologia , Células Dendríticas/imunologia , Feminino , Citometria de Fluxo , Técnica Indireta de Fluorescência para Anticorpo , Imunidade Inata , Células Matadoras Naturais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA