Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Glia ; 66(12): 2700-2718, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30277607

RESUMO

Emerging evidence have posited that dysregulated microglia impair clearance and containment of amyloid-ß (Aß) species in the brain, resulting in aberrant buildup of Aß and onset of Alzheimer's disease (AD). Hematopoietic cell kinase (Hck) is one of the key regulators of phagocytosis among the Src family tyrosine kinases (SFKs) in myeloid cells, and its expression is found to be significantly altered in AD brains. However, the role of Hck signaling in AD pathogenesis is unknown. We employed pharmacological inhibition and genetic ablation of Hck in BV2 microglial cells and J20 mouse model of AD, respectively, to evaluate the impact of Hck deficiency on Aß-stimulated microglial phagocytosis, Aß clearance, and resultant AD-like neuropathology. Our in vitro data reveal that pharmacological inhibition of SFKs/Hck in BV2 cells and genetic ablation of their downstream kinase, spleen tyrosine kinase (Syk), in primary microglia significantly attenuate Aß oligomers-stimulated microglial phagocytosis. Whereas in Hck-deficient J20 mice, we observed exacerbated Aß plaque burden, reduced microglial coverage, containment, and phagocytosis of Aß plaques, and induced iNOS expression in plaque-associated microglial clusters. These multifactorial changes in microglial activities led to attenuated PSD95 levels in hippocampal DG and CA3 regions, but did not alter the postsynaptic dendritic spine morphology at the CA1 region nor cognitive function of the mice. Hck inhibition thus accelerates early stage AD-like neuropathology by dysregulating microglial function and inducing neuroinflammation. Our data implicate that Hck pathway plays a prominent role in regulating microglial neuroprotective function during the early stage of AD development.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Regulação da Expressão Gênica/genética , Microglia/enzimologia , Proteínas Proto-Oncogênicas c-hck/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetulus , Modelos Animais de Doenças , Antagonistas de Estrogênios/farmacologia , Comportamento Exploratório/fisiologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/ultraestrutura , Fagocitose/efeitos dos fármacos , Fagocitose/genética , Proteínas Proto-Oncogênicas c-hck/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Quinase Syk/genética , Quinase Syk/metabolismo , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Transfecção
2.
Proc Natl Acad Sci U S A ; 109(34): 13757-62, 2012 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-22875704

RESUMO

The rs1061170T/C variant encoding the Y402H change in complement factor H (CFH) has been identified by genome-wide association studies as being significantly associated with age-related macular degeneration (AMD). However, the precise mechanism by which this CFH variant impacts the risk of AMD remains largely unknown. Oxidative stress plays an important role in many aging diseases, including cardiovascular disease and AMD. A large amount of oxidized phospholipids (oxPLs) are generated in the eye because of sunlight exposure and high oxygen content. OxPLs bind to the retinal pigment epithelium and macrophages and strongly activate downstream inflammatory cascades. We hypothesize that CFH may impact the risk of AMD by modulating oxidative stress. Here we demonstrate that CFH binds to oxPLs. The CFH 402Y variant of the protective rs1061170 genotype binds oxPLs with a higher affinity and exhibits a stronger inhibitory effect on the binding of oxPLs to retinal pigment epithelium and macrophages. In addition, plasma from non-AMD subjects with the protective genotype has a lower level of systemic oxidative stress measured by oxPLs per apolipoprotein B (oxPLs/apoB). We also show that oxPL stimulation increases expression of genes involved in macrophage infiltration, inflammation, and neovascularization in the eye. OxPLs colocalize with CFH in drusen in the human AMD eye. Subretinal injection of oxPLs induces choroidal neovascularization in mice. In addition, we show that the CFH risk allele confers higher complement activation and cell lysis activity. Together, these findings suggest that CFH influences AMD risk by modulating oxidative stress, inflammation, and abnormal angiogenesis.


Assuntos
Fator H do Complemento/genética , Degeneração Macular/genética , Fosfolipídeos/química , Idoso de 80 Anos ou mais , Angiografia/métodos , Animais , Genótipo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Drusas do Disco Óptico/metabolismo , Oxigênio/química
3.
J Biol Chem ; 287(2): 1520-6, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22049084

RESUMO

Genome-wide association study (GWAS) has identified genetic variants in the promoter region of the high temperature requirement factor A1 (HTRA1) gene associated with age-related macular degeneration (AMD). As a secreted serine protease, HTRA1 has been reported to interact with members of the transforming growth factor-ß (TGF-ß) family and regulate their signaling pathways. Growth differentiation factor 6 (GDF6), a member of the TGF-ß family, is involved in ectoderm patterning and eye development. Mutations in GDF6 have been associated with abnormal eye development that may result in microphthalmia and anophthalmia. In this report, we identified a single nucleotide polymorphism (SNP) rs6982567 A/G near the GDF6 gene that is significantly associated with AMD (p value = 3.54 × 10(-8)). We demonstrated that the GDF6 AMD risk allele (rs6982567 A) is associated with decreased expression of the GDF6 and increased expression of HTRA1. Similarly, the HTRA1 AMD risk allele (rs10490924 T) is associated with decreased GDF6 and increased HTRA1 expression. We observed decreased vascular development in the retina and significant up-regulation of GDF6 gene in the RPE layer, retinal and brain tissues in HTRA1 knock-out (htra1(-/-)) mice as compared with the wild-type counterparts. Furthermore, we showed enhanced SMAD signaling in htra1(-/-) mice. Our data suggests a critical role of HTRA1 in the regulation of angiogenesis via TGF-ß signaling and identified GDF6 as a novel disease gene for AMD.


Assuntos
Fator 6 de Diferenciação de Crescimento/biossíntese , Degeneração Macular/metabolismo , Neovascularização Patológica/metabolismo , Polimorfismo de Nucleotídeo Único , Serina Endopeptidases/biossíntese , Idoso , Alelos , Animais , Estudos de Coortes , Feminino , Regulação da Expressão Gênica/genética , Fator 6 de Diferenciação de Crescimento/genética , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Degeneração Macular/genética , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Retina/metabolismo , Retina/patologia , Fatores de Risco , Serina Endopeptidases/genética , Transdução de Sinais/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
4.
Toxicol Sci ; 193(2): 175-191, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37074955

RESUMO

Exposure to traffic-related air pollution consisting of particulate matter (PM) is associated with cognitive decline leading to Alzheimer's disease (AD). In this study, we sought to examine the neurotoxic effects of exposure to ultrafine PM and how it exacerbates neuronal loss and AD-like neuropathology in wildtype (WT) mice and a knock-in mouse model of AD (AppNL-G-F/+-KI) when the exposure occurs at a prepathologic stage or at a later age with the presence of neuropathology. AppNL-G-F/+-KI and WT mice were exposed to concentrated ultrafine PM from local ambient air in Irvine, California, for 12 weeks, starting at 3 or 9 months of age. Particulate matter-exposed animals received concentrated ultrafine PM up to 8 times above the ambient levels, whereas control animals were exposed to purified air. Particulate matter exposure resulted in a marked impairment of memory tasks in prepathologic AppNL-G-F/+-KI mice without measurable changes in amyloid-ß pathology, synaptic degeneration, and neuroinflammation. At aged, both WT and AppNL-G-F/+-KI mice exposed to PM showed a significant memory impairment along with neuronal loss. In AppNL-G-F/+-KI mice, we also detected an increased amyloid-ß buildup and potentially harmful glial activation including ferritin-positive microglia and C3-positive astrocytes. Such glial activation could promote the cascade of degenerative consequences in the brain. Our results suggest that exposure to PM impairs cognitive function at both ages while exacerbation of AD-related pathology and neuronal loss may depend on the stage of pathology, aging, and/or state of glial activation. Further studies will be required to unveil the neurotoxic role of glial activation activated by PM exposure.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/patologia , Material Particulado/toxicidade , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças , Encéfalo/metabolismo , Transtornos da Memória/induzido quimicamente , Camundongos Transgênicos
5.
J Trace Elem Med Biol ; 62: 126578, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32599538

RESUMO

BACKGROUND: Copper (Cu) is an essential metal mediating a variety of vital biological reactions with its redox property. Its dyshomeostasis has been associated with accelerated cognitive decline and neurodegenerative disorders, such as Alzheimer's disease (AD). However, underlying neurotoxic mechanisms elicited by dysregulated Cu remain largely elusive. We and others previously demonstrated that exposure to Cu in drinking water significantly exacerbated pathological hallmarks of AD and pro-inflammatory activation of microglia, coupled with impaired phagocytic capacity, in mouse models of AD. METHODS: In the present study, we extended our investigation to evaluate whether chronic Cu exposure to wild-type (WT) and J20 mouse model of AD perturbs homeostatic dynamics of microglia and contributes to accelerated transformation of microglia towards degenerative phenotypes that are closely associated with neurodegeneration. We further looked for evidence of alterations in the microglial morphology and spatial memory of the Cu-exposed mice to assess the extent of the Cu toxicity. RESULTS: We find that chronic Cu exposure to pre-pathological J20 mice upregulates the translation of degenerative genes and represses homeostatic genes within microglia even in the absence amyloid-beta plaques. We also observe similar expression signatures in Cu-exposed WT mice, suggesting that excess Cu exposure alone could lead to perturbed microglial homeostatic phenotypes and contribute to accelerated cognitive decline. CONCLUSION: Our findings highlight the risk of chronic Cu exposure on cognitive decline and altered microglia activation towards degenerative phenotypes. These changes may represent one of the key mechanisms linking Cu exposure or its dyshomeostasis to an increased risk for AD.


Assuntos
Doença de Alzheimer/etiologia , Transtornos Cognitivos/induzido quimicamente , Cobre/toxicidade , Microglia/efeitos dos fármacos , Microglia/patologia , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/genética , Animais , Transtornos Cognitivos/patologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamoxifeno/farmacologia , Testes de Toxicidade Crônica
6.
Mol Neurobiol ; 57(5): 2447-2460, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32146679

RESUMO

Microglial dysregulation, pertaining to impairment in phagocytosis, clearance and containment of amyloid-ß (Aß), and activation of neuroinflammation, has been posited to contribute to the pathogenesis of Alzheimer's disease (AD). Detailed cellular mechanisms that are disrupted during the disease course to display such impairment in microglia, however, remain largely undetermined. We hypothesize that loss of hematopoietic cell kinase (HCK), a phagocytosis-regulating member of the Src family tyrosine kinases that mediate signals from triggering receptor expressed on myeloid cells 2 and other immunoreceptors, impairs microglial homeostasis and Aß clearance, leading to the accelerated buildup of Aß pathology and cognitive decline during the early stage of neuropathological development. To elucidate the pivotal role of HCK in AD, we generated a constitutive knockout of HCK in the Tg2576 mouse model of AD. We found that HCK deficiency accelerated cognitive decline along with elevated Aß level and plaque burden, attenuated microglial Aß phagocytosis, induced iNOS expression in microglial clusters, and reduced pre-synaptic protein at the hippocampal regions. Our findings substantiate that HCK plays a prominent role in regulating microglial neuroprotective functions and attenuating early AD neuropathology.


Assuntos
Doença de Alzheimer/enzimologia , Microglia/enzimologia , Proteínas Proto-Oncogênicas c-hck/deficiência , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Progressão da Doença , Comportamento Exploratório , Feminino , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microglia/patologia , Teste do Labirinto Aquático de Morris , Células Mieloides/enzimologia , Neuroimunomodulação , Fagocitose , Placa Amiloide , Proteínas Proto-Oncogênicas c-hck/genética , Reconhecimento Psicológico
7.
Toxicol Sci ; 170(1): 144-156, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30923833

RESUMO

Chronic exposure to copper and its dyshomeostasis have been linked to accelerated cognitive decline and potentially increasing risk for Alzheimer's disease (AD). We and others have previously demonstrated that exposure to copper through drinking water significantly increased parenchymal amyloid-beta (Aß) plaques and decreased endothelial low-density lipoprotein receptor-related protein 1 (LRP1) in mouse models of AD. In this study, we determined the underlying mechanisms that microRNA critically mediated the copper-induced loss of endothelial LRP1. In human primary microvascular endothelial cells (MVECs), microRNA-200b-3p, -200c-3p, and -205-5p were significantly elevated within the 24-h exposure to copper and returned to baseline after 48-h postexposure, which corresponded with the temporal change of LRP1 expression in these cells. Transient expression of synthetic microRNA-200b-3p, -200c-3p, or -205-5p on MVECs significantly decreased endothelial LRP1, and cotreatment of synthetic antagomirs effectively prevented the loss of LRP1 during copper exposure, collectively supporting the key regulatory role of these microRNAs in copper-induced loss of LRP1. In mice, a significant reduction of LRP1 in cortical vasculature was evident following 9 months exposure to 1.3 ppm copper in drinking water, although the levels of cortical microRNA-205-5p, -200b-3p, and -200c-3p were only marginally elevated. This, however, correlated with increased vascular accumulation of Aß and impairment of spatial memory, indicating that copper exposure has the pivotal role in the vascular damage and development of cognitive decline.


Assuntos
Doença de Alzheimer/induzido quimicamente , Encéfalo/efeitos dos fármacos , Cobre/toxicidade , Células Endoteliais/efeitos dos fármacos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , MicroRNAs/genética , Doença de Alzheimer/metabolismo , Animais , Encéfalo/irrigação sanguínea , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Memória Espacial/efeitos dos fármacos , Transfecção , Regulação para Cima
8.
Int J Pharm ; 307(1): 42-50, 2006 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-16260103

RESUMO

The aim of this study was to correlate the taxonomy of grapefruit, pummelo, orange, lime and lemon with fruit juice-mediated cytotoxicity, modulation of epithelial permeability and P-glycoprotein (P-gp)-mediated efflux using 0-50% juice concentrations. Lime and lemon juices at 30% enhanced the absorption of [14C]-mannitol across Caco-2 cell monolayers by six- and eight-fold, respectively, but grapefruit and pummelo juices did not modulate the paracellular [14C]-mannitol transport even at 50%. Orange juice at 30% increased mannitol absorption to a comparable level as lime juice, but had minimal effects on TEER. All five juices did not modulate the passive diffusional pathway as exemplified by their negligible effects on [3H]-propranolol absorption. Grapefruit, pummelo and orange juices showed P-gp inhibitory activity by reducing rhodamine-123 (R-123) efflux and elevating R-123 cellular accumulation, but lime and lemon juices did not. Lime and lemon juices at >or=30% were cytotoxic towards Caco-2 cells. Grapefruit and pummelo juices at 10% did not affect Caco-2 cell viability, but they enhanced cell growth at concentrations of >or=30%. Orange juice increased cell viability only at lower concentrations. On the basis of these data, lime and lemon juices could be regarded as a group distinct from grapefruit and pummelo juices, while orange juice appeared to belong to a bridging group. This grouping was consistent with the categorization of the citrus fruits according to their dominant flavonoid pattern and taxonomy.


Assuntos
Bebidas , Citrus , Interações Alimento-Droga , Absorção , Bebidas/toxicidade , Transporte Biológico/efeitos dos fármacos , Células CACO-2 , Radioisótopos de Carbono , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citrus/classificação , Humanos , Manitol/metabolismo , Propranolol/metabolismo
9.
Microbes Infect ; 17(8): 549-56, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25912134

RESUMO

Alzheimer's disease (AD) is a leading cause of dementia among elderly. Yet, its etiology remains largely unclear. In this review, we summarize studies that associate systemic infection and neuroinflammation with AD, while highlighting that early-life or life-long exposure to infectious agents predisposes one to develop AD at a later age.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/complicações , Humanos , Infecções/complicações , Infecções/patologia , Inflamação/complicações , Inflamação/patologia , Fatores de Risco
10.
Neurobiol Aging ; 36(7): 2260-2271, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25964214

RESUMO

Glial glutamate transporter, GLT-1, is the major Na(+)-driven glutamate transporter to control glutamate levels in synapses and prevent glutamate-induced excitotoxicity implicated in neurodegenerative disorders including Alzheimer's disease (AD). Significant functional loss of GLT-1 has been reported to correlate well with synaptic degeneration and severity of cognitive impairment among AD patients, yet the underlying molecular mechanism and its pathological consequence in AD are not well understood. Here, we find the temporal decrease in GLT-1 levels in the hippocampus of the 3xTg-AD mouse model and that the pharmacological upregulation of GLT-1 significantly ameliorates the age-dependent pathological tau accumulation, restores synaptic proteins, and rescues cognitive decline with minimal effects on Aß pathology. In primary neuron and astrocyte coculture, naturally secreted Aß species significantly downregulate GLT-1 steady-state and expression levels. Taken together, our data strongly suggest that GLT-1 restoration is neuroprotective and Aß-induced astrocyte dysfunction represented by a functional loss of GLT-1 may serve as one of the major pathological links between Aß and tau pathology.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Ceftriaxona/farmacologia , Ceftriaxona/uso terapêutico , Transportador 2 de Aminoácido Excitatório/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Células Cultivadas , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Ácido Glutâmico/toxicidade , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia , Regulação para Cima/efeitos dos fármacos
11.
Semin Ophthalmol ; 26(3): 216-24, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21609235

RESUMO

There is currently no FDA-approved therapy for treating patients with geographic atrophy (GA), a late stage of age-related macular degeneration (AMD). Cell transplantation has the potential to restore vision in these patients. This review discusses how recent advancement in induced pluripotent stem (iPS) cells provides a promising therapy for GA treatment. Recent advances in stem cell biology have demonstrated that it is possible to derive iPS cells from human somatic cells by introducing reprogramming factors. Human retinal pigment epithelium (RPE) cells and photoreceptors can be derived from iPS cells by defined factors. Studies show that transplanting these cells can stabilize or recover vision in animal models. However, cell derivation protocols and transplantation procedures still need to be optimized. Much validation has to be done before clinical-grade, patient-derived iPS can be applied for human therapy. For now, RPE cells and photoreceptors derived from patient-specific iPS cells can serve as a valuable tool in elucidating the mechanism of pathogenesis and drug discovery for GA.


Assuntos
Atrofia Geográfica/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Degeneração Macular/terapia , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Humanos
12.
Expert Opin Drug Metab Toxicol ; 5(7): 789-801, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19545213

RESUMO

Food-drug interactions have been associated with clinically important pharmacokinetic and pharmacodynamic changes of a drug. The aim of this paper is to review the regulation of P-glycoprotein (P-gp) by dietary components and to correlate the changes in cellular P-gp function and expression with drug bioavailability. In summary, the published literature has provided extensive data supporting the modulation of drug bioavailability through P-gp regulation by components in food groups such as fruit juices, spices, herbs, cruciferous vegetables and green tea. Most of these data were, however, derived from in vitro cell models and, except for the St John's wort, the clinical significance of most reported interactions remains to be clarified. Studies on piperine and capsaicin have underscored an often poor correlation between in vivo and in vitro data, whereas experiments involving curcumin highlighted differences between acute and chronic consumption of a dietary component on P-gp function and expression in vivo. A better understanding of the pharmacokinetic and pharmacodynamic profiles of the dietary components will aid in addressing these knowledge gaps.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Dieta , Interações Alimento-Droga/fisiologia , Regulação da Expressão Gênica/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Interações Ervas-Drogas/fisiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA