Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Brain Behav Immun ; 65: 262-273, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28526436

RESUMO

Neuroinflammation is a pervasive feature of Alzheimer's disease (AD) and characterized by activated microglia, increased proinflammatory cytokines and/or infiltrating immune cells. T helper 17 (Th17) cells are found in AD brain parenchyma and interleukin-17A (IL-17A) is identified around deposits of aggregated amyloid ß protein (Aß). However, the role of IL-17A in AD pathogenesis remains elusive. We overexpressed IL-17A in an AD mouse model via recombinant adeno-associated virus serotype 5 (rAAV5)-mediated intracranial gene delivery. AD model mice subjected to injection of a vehicle (PBS) or rAAV5 carrying the lacZ gene served as controls. IL-17A did not exacerbate neuroinflammation in IL-17A-overexpressing mice. We found that IL-17A overexpression markedly improved glucose metabolism, decreased soluble Aß levels in the hippocampus and cerebrospinal fluid, drastically reduced cerebral amyloid angiopathy, and modestly but significantly improved anxiety and learning deficits. Moreover, the ATP-binding cassette subfamily A member 1 (ABCA1), which can transport Aß from the brain into the blood circulation, significantly increased in IL-17A-overexpressing mice. In vitro treatment of brain endothelial bEnd.3 cells with IL-17A induced a dose-dependent increase in protein expression of ABCA1 through ERK activation. Our study suggests that IL-17A may decrease Aß levels in the brain by upregulating ABCA1 in blood-brain barrier endothelial cells.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Angiopatia Amiloide Cerebral/genética , Interleucina-17/farmacologia , Transportador 1 de Cassete de Ligação de ATP/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Microglia/metabolismo , Células Th17/metabolismo , Ativação Transcricional , Regulação para Cima
2.
Inflamm Res ; 66(11): 993-997, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28681194

RESUMO

OBJECTIVE AND DESIGN: Work in multiple organs has suggested that toll-like receptor 4 (TLR4) may play a role in insulin resistance. Additional studies have shown a negative role for TLR4 on retinal health. We have previously reported that ß-adrenergic receptors can regulate both TLR4 signal transduction, as well as insulin signaling in the retina and in retinal endothelial cells. Thus, we hypothesized that TLR4 would regulate retinal insulin signaling. MATERIALS AND METHODS: We used endothelial cell-specific TLR4 knockout mice, as well as TLR4-overexpressing mice for these studies. METHODS: Western blotting and ELISA analyses were done for investigations of insulin receptor, insulin receptor substrate 1 (IRS-1) serine 307, and Akt phosphorylation, as well as cleaved caspase 3 levels in the mouse retina. RESULTS: We found that loss of TLR4 led to increased insulin receptor and Akt phosphorylation, as well as decreased IRS-1Ser307 levels. In support of these results, TLR4 overexpression decreased insulin signaling and the cleavage of caspase 3. CONCLUSIONS: Therefore, these results suggest that TLR4 plays a key role in insulin signaling in the retina. Reduction of TLR4 levels may be protective to the retina.


Assuntos
Resistência à Insulina/fisiologia , Retina/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Apoptose , Camundongos Transgênicos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Receptor 4 Toll-Like/genética
3.
J Biol Chem ; 290(16): 10229-41, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25724648

RESUMO

Classical immunization methods do not generate catalytic antibodies (catabodies), but recent findings suggest that the innate antibody repertoire is a rich catabody source. We describe the specificity and amyloid ß (Aß)-clearing effect of a catabody construct engineered from innate immunity principles. The catabody recognized the Aß C terminus noncovalently and hydrolyzed Aß rapidly, with no reactivity to the Aß precursor protein, transthyretin amyloid aggregates, or irrelevant proteins containing the catabody-sensitive Aß dipeptide unit. The catabody dissolved preformed Aß aggregates and inhibited Aß aggregation more potently than an Aß-binding IgG. Intravenous catabody treatment reduced brain Aß deposits in a mouse Alzheimer disease model without inducing microgliosis or microhemorrhages. Specific Aß hydrolysis appears to be an innate immune function that could be applied for therapeutic Aß removal.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Anticorpos Catalíticos/metabolismo , Encéfalo/metabolismo , Anticorpos de Cadeia Única/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Animais , Anticorpos Catalíticos/química , Anticorpos Catalíticos/genética , Encéfalo/imunologia , Encéfalo/patologia , Modelos Animais de Doenças , Expressão Gênica , Células HEK293 , Humanos , Hidrólise , Imunidade Inata , Camundongos , Fragmentos de Peptídeos/química , Engenharia de Proteínas , Proteólise , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética
4.
Biochem Biophys Res Commun ; 479(2): 331-337, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27641666

RESUMO

Microglia-mediated clearance of amyloid beta-protein (Aß) via Toll-like receptor 4 (TLR4) signaling may play an important role in the pathogenesis of Alzheimer's disease (AD). However, as the disease progresses, activated microglia appear to become incapable of clearing Aß deposits. Because repeated exposure to a TLR4 ligand leads to a diminished response of monocytes/macrophages to lipopolysaccharide (LPS) and because aggregated Aß is a TLR4 ligand, we hypothesize that chronic exposure of microglia to Aß deposits may induce a state of Toll-like receptor (TLR) signaling dysfunction, leading to decreased Aß clearance and accelerated disease progression. LPS or phosphate-buffered saline (PBS) was injected into the hippocampus of AD-model (TgAPP/PS1) and wild-type (non-Tg) mice before and after the onset of Aß deposition, at age 2 and 12 months, respectively. Brain specimens were collected 7 days post-injection and analyzed for microglial activation and Aß load. While LPS-injected 2-month-old non-Tg mice showed 48-fold and 11-fold greater Iba1 immunoreactivity in the neocortex and hippocampus, respectively, compared with PBS-injected mice, LPS-injected 2-month-old TgAPP/PS1 mice had 61-fold and 13-fold increases in the neocortex and hippocampus, respectively. LPS injection activated microglia more strongly in TgAPP/PS1 mice than in non-Tg mice at 2 months of age. In contrast, at 12 months of age, Iba1 immunoreactivity of microglia was increased 541-fold and 38-fold in the neocortex and hippocampus, respectively, in LPS-injected non-Tg mice and 2.7-fold and 3.3-fold in the neocortex and hippocampus, respectively, in LPS-injected TgAPP/PS1 mice. Surprisingly, LPS injection decreased CD45 immunoreactivity in TgAPP/PS1 mice but increased it in non-Tg mice at 12 months. Although microglia in 12-month-old non-Tg mice showed stronger response to LPS than 2-month-old non-Tg mice, microglia in TgAPP/PS1 mice exhibited diminished immune response to LPS during aging. Our data indicate that microglial TLR4 signaling is altered in an AD mouse model and suggest that altered TLR4 signaling may contribute to Aß accumulation in the brain.


Assuntos
Envelhecimento , Doença de Alzheimer/genética , Lipopolissacarídeos/química , Microglia/metabolismo , Receptor 4 Toll-Like/genética , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Hipocampo/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Monócitos/metabolismo , Transdução de Sinais
5.
Brain Behav Immun ; 53: 84-95, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26562537

RESUMO

Interleukin-17A (IL-17A) is generally considered as one of the pathogenic factors involved in multiple sclerosis (MS). Indirect evidence for this is that IL-17A-producing T helper 17 (Th17) cells preferentially accumulate in lesions of MS and experimental autoimmune encephalomyelitis (EAE). However, a direct involvement of IL-17A in MS pathogenesis is still an open question. In this study, we overexpressed IL-17A in the brains of mice (IL-17A-in-Brain mice) via recombinant adeno-associated virus serotype 5 (rAAV5)-mediated gene delivery. In spite of high levels of IL-17A expression in the brain and blood, IL-17A-in-Brain mice exhibit no inflammatory responses and no abnormalities in motor coordination and spatial orientation. Unexpectedly, IL-17A-in-Brain mice show decreases in body weight and adipose tissue mass and an improvement in glucose tolerance and insulin sensitivity. IL-17A enhances glucose uptake in PC12 cells by activation of AKT. Our results provide direct evidence for the first time that IL-17A overexpression in the central nervous system does not cause physical and learning disabilities and neuroinflammation and suggest that IL-17A may regulate glucose metabolism through the AKT signaling pathway.


Assuntos
Encefalomielite Autoimune Experimental/etiologia , Glucose/metabolismo , Interleucina-17/administração & dosagem , Deficiências da Aprendizagem/etiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tecido Adiposo/patologia , Animais , Citocinas/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Técnicas de Transferência de Genes , Resistência à Insulina , Interleucina-17/biossíntese , Interleucina-17/genética , Deficiências da Aprendizagem/metabolismo , Deficiências da Aprendizagem/patologia , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Células PC12 , Ratos , Transdução de Sinais , Células Th17/imunologia , Células Th17/metabolismo
6.
PLoS One ; 18(7): e0288497, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440496

RESUMO

In order to study effects of macrophage-derived inflammatory mediators associated with systemic inflammation on brain endothelial cells, we have established a co-culture system consisting of bEnd.3 cells and LPS-activated Raw 264.7 cells and performed its cytokine profiling. The cytokine profile of the co-culture model was compared to that of mice treated with intraperitoneal LPS injection. We found that, among cytokines profiled, eight cytokines/chemokines were similarly upregulated in both in vivo mouse and in vitro co-culture model. In contrast to the co-culture model, the cytokine profile of a common mono-culture system consisting of only LPS-activated bEnd.3 cells had little similarity to that of the in vivo mouse model. These results indicate that the co-culture of bEnd.3 cells with LPS-activated Raw 264.7 cells is a better model than the common mono-culture of LPS-activated bEnd.3 cells to investigate the molecular mechanism in endothelial cells, by which systemic inflammation induces neuroinflammation. Moreover, fibrinogen adherence both to bEnd.3 cells in the co-culture and to brain blood vessels in a LPS-treated animal model of Alzheimer's disease increased. To the best of our knowledge, this is the first to utilize bEnd.3 cells co-cultured with LPS-activated Raw 264.7 cells as an in vitro model to investigate the consequence of macrophage-derived inflammatory mediators on brain endothelial cells.


Assuntos
Citocinas , Células Endoteliais , Animais , Camundongos , Citocinas/metabolismo , Células Endoteliais/metabolismo , Técnicas de Cocultura , Lipopolissacarídeos/efeitos adversos , Encéfalo/metabolismo , Macrófagos/metabolismo , Inflamação/induzido quimicamente , Mediadores da Inflamação
7.
Rev Neurosci ; 23(4): 363-79, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23089603

RESUMO

The discovery of gene mutations underlying autosomal dominant Alzheimer's disease has enabled researchers to reproduce several hallmarks of this disorder in transgenic mice, notably the formation of Aß plaques in brain and cognitive deficits. APP transgenic mutants have also been investigated with respect to survival rates, neurologic functions, and motor coordination, which are all susceptible to alteration in Alzheimer dementia. Several transgenic lines expressing human mutated or wild-type APP had higher mortality rates than non-transgenic controls with or without the presence of Aß plaques. Mortality rates were also elevated in APP transgenic mice with vascular amyloid accumulation, thereby implicating cerebrovascular factors in the precocious death observed in all APP transgenic models. In addition, myoclonic jumping has been described in APP mutants, together with seizure activity, abnormal limb-flexion and paw-clasping reflexes, and motor coordination deficits. The neurologic signs resemble the myoclonic movements, epileptic seizures, pathological reflexes, and gait problems observed in late-stage Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Predisposição Genética para Doença/genética , Transtornos dos Movimentos/genética , Doenças do Sistema Nervoso/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Transtornos dos Movimentos/fisiopatologia , Mutação/genética , Doenças do Sistema Nervoso/fisiopatologia
8.
Am J Pathol ; 179(3): 1095-103, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21763676

RESUMO

The accumulation of ß-amyloid protein (Aß) in the brain is thought to be a primary etiologic event in Alzheimer's disease (AD). Fibrillar Aß plaques, a hallmark of AD abnormality, are closely associated with activated microglia. Activated microglia have contradictory roles in the pathogenesis of AD, being either neuroprotective (by clearing harmful Aß and repairing damaged tissues) or neurotoxic (by producing proinflammatory cytokines and reactive oxygen species). Aß aggregates can activate microglia by interacting with multiple toll-like receptors (TLRs), the pattern-recognition receptors of the innate immune system. Because the adapter protein MyD88 is essential for the downstream signaling of all TLRs, except TLR3, we investigated the effects of MyD88 deficiency (MyD88(-/-)) on Aß accumulation and microglial activation in an AD mouse model. MyD88 deficiency decreased Aß load and microglial activation in the brain. The decrease in Aß load in an MyD88(-/-) AD mouse model was associated with increased and decreased protein expression of apolipoprotein E (apoE) and CX3CR1, respectively, compared with that in an MyD88 wild-type AD mouse model. These results suggest that MyD88 deficiency may reduce Aß load by enhancing the phagocytic capability of microglia through fractalkine (the ligand of CX3CR1) signaling and by promoting apoE-mediated clearance of Aß from the brain. These findings also suggest that chronic inflammatory responses induced by Aß accumulation via the MyD88-dependent signaling pathway exacerbate ß-amyloidosis in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/deficiência , Animais , Apolipoproteínas E/metabolismo , Receptor 1 de Quimiocina CX3C , Cérebro/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Quimiocinas/metabolismo
9.
J Neuroinflammation ; 8: 92, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21827663

RESUMO

BACKGROUND: Amyloid plaques, a pathological hallmark of Alzheimer's disease (AD), are accompanied by activated microglia. The role of activated microglia in the pathogenesis of AD remains controversial: either clearing Aß deposits by phagocytosis or releasing proinflammatory cytokines and cytotoxic substances. Microglia can be activated via toll-like receptors (TLRs), a class of pattern-recognition receptors in the innate immune system. We previously demonstrated that an AD mouse model homozygous for a loss-of-function mutation of TLR4 had increases in Aß deposits and buffer-soluble Aß in the brain as compared with a TLR4 wild-type AD mouse model at 14-16 months of age. However, it is unknown if TLR4 signaling is involved in initiation of Aß deposition as well as activation and recruitment of microglia at the early stage of AD. Here, we investigated the role of TLR4 signaling and microglial activation in early stages using 5-month-old AD mouse models when Aß deposits start. METHODS: Microglial activation and amyloid deposition in the brain were determined by immunohistochemistry in the AD models. Levels of cerebral soluble Aß were determined by ELISA. mRNA levels of cytokines and chemokines in the brain and Aß-stimulated monocytes were quantified by real-time PCR. Cognitive functions were assessed by the Morris water maze. RESULTS: While no difference was found in cerebral Aß load between AD mouse models at 5 months with and without TLR4 mutation, microglial activation in a TLR4 mutant AD model (TLR4M Tg) was less than that in a TLR4 wild-type AD model (TLR4W Tg). At 9 months, TLR4M Tg mice had increased Aß deposition and soluble Aß42 in the brain, which were associated with decrements in cognitive functions and expression levels of IL-1ß, CCL3, and CCL4 in the hippocampus compared to TLR4W Tg mice. TLR4 mutation diminished Aß-induced IL-1ß, CCL3, and CCL4 expression in monocytes. CONCLUSION: This is the first demonstration of TLR4-dependent activation of microglia at the early stage of ß-amyloidosis. Our results indicate that TLR4 is not involved in the initiation of Aß deposition and that, as Aß deposits start, microglia are activated via TLR4 signaling to reduce Aß deposits and preserve cognitive functions from Aß-mediated neurotoxicity.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Transtornos Cognitivos/fisiopatologia , Microglia/fisiologia , Receptor 4 Toll-Like/genética , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Comportamento Animal/fisiologia , Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Microglia/citologia , Mutação , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
10.
J Mol Neurosci ; 71(6): 1245-1255, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33185814

RESUMO

Repeated exposure to toll-like receptor 4 (TLR4) ligands, such as lipopolysaccharide (LPS), reduces responses of monocytes/macrophages to LPS (LPS/endotoxin tolerance). Microglial exposure to Aß deposits, a TLR4 ligand, may cause "Aß/LPS tolerance," leading to decreased Aß clearance. We demonstrated that microglial activation by LPS is diminished in Aß deposit-bearing 12-month-old model mice of Alzheimer's disease (AD), compared with non-AD mice and Aß deposit-free 2-month-old AD mice. Because miR-146a plays a predominant role in inducing TLR tolerance in macrophages and because miR-146a in extracellular vesicles (EVs) shed by inflammatory macrophages increases in circulation, we investigated potential roles of miR-146a and inflammatory EVs in inducing TLR tolerance in microglia and in altering expression of inflammatory AD risk genes. We found that miR-146a upregulation induces TLR tolerance and alters expression of inflammatory AD risk genes in response to LPS treatment in BV2 microglia. LPS brain injection altered expression of the AD risk genes in 12-month-old AD mice but not in non-AD littermates. EVs from inflammatory macrophages polarize BV2 microglia to M1 phenotype and induce TLR tolerance. Microglia exposed to Aß in the brain show reduced cytokine responses to systemic inflammation due to peripheral LPS injection, indicating TLR/Aß tolerance in microglia. Our results suggest that increased miR-146a induces microglial Aß/LPS tolerance and that circulating EVs shed by inflammatory macrophages contribute to microglial Aß/LPS tolerance, leading to reduced Aß clearance. Our study also suggests that altered expression of inflammatory AD risk genes may contribute to AD development via the same molecular mechanism underlying LPS tolerance.


Assuntos
Doença de Alzheimer/genética , Exossomos/genética , MicroRNAs/metabolismo , Receptor 4 Toll-Like/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/toxicidade , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Exossomos/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Microglia/efeitos dos fármacos , Microglia/metabolismo , Presenilina-1/genética , Células RAW 264.7 , Transdução de Sinais
11.
Int J Endocrinol ; 2020: 6942307, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33376487

RESUMO

Obesity significantly increases the risk of developing type 2 diabetes mellitus and other metabolic diseases. Obesity is associated with chronic low-grade inflammation in white adipose tissues, which is thought to play an essential role in developing insulin resistance. Many lines of evidence indicate that toll-like receptors (TLRs) and their downstream signaling pathways are involved in development of chronic low-grade inflammation and insulin resistance, which are associated with obesity. Mice lacking molecules positively involved in the TLR signaling pathways are generally protected from high-fat diet-induced inflammation and insulin resistance. In this study, we have determined the effects of genetic deficiency of toll/interleukin-1 receptor-domain-containing adaptor-inducing interferon-ß (TRIF) on food intake, bodyweight, glucose metabolism, adipose tissue macrophage polarization, and insulin signaling in normal chow diet-fed mice to investigate the role of the TRIF-dependent TLR signaling in adipose tissue metabolism and inflammation. TRIF deficiency (TRIF-/-) increased food intake and bodyweight. The significant increase in bodyweight in TRIF-/- mice was discernible as early as 24 weeks of age and sustained thereafter. TRIF-/- mice showed impaired glucose tolerance in glucose tolerance tests, but their insulin tolerance tests were similar to those in TRIF+/+ mice. Although no difference was found in the epididymal adipose mass between the two groups, the percentage of CD206+ M2 macrophages in epididymal adipose tissue decreased in TRIF-/- mice compared with those in TRIF+/+ mice. Furthermore, activation of epididymal adipose AKT in response to insulin stimulation was remarkably diminished in TRIF-/- mice compared with TRIF+/+ mice. Our results indicate that the TRIF-dependent TLR signaling contributes to maintaining insulin/AKT signaling and M2 macrophages in epididymal adipose tissue under a normal chow diet and provide new evidence that TLR4-targeted therapies for type 2 diabetes require caution.

12.
Front Immunol ; 11: 724, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32391019

RESUMO

Amyloid plaques, mainly composed of abnormally aggregated amyloid ß-protein (Aß) in the brain parenchyma, and neurofibrillary tangles (NFTs), consisting of hyperphosphorylated tau protein aggregates in neurons, are two pathological hallmarks of Alzheimer's disease (AD). Aß fibrils and tau aggregates in the brain are closely associated with neuroinflammation and synapse loss, characterized by activated microglia and dystrophic neurites. Genome-wide genetic association studies revealed important roles of innate immune cells in the pathogenesis of late-onset AD by recognizing a dozen genetic risk loci that modulate innate immune activities. Furthermore, microglia, brain resident innate immune cells, have been increasingly recognized to play key, opposing roles in AD pathogenesis by either eliminating toxic Aß aggregates and enhancing neuronal plasticity or producing proinflammatory cytokines, reactive oxygen species, and synaptotoxicity. Aggregated Aß binds to toll-like receptor 4 (TLR4) and activates microglia, resulting in increased phagocytosis and cytokine production. Complement components are associated with amyloid plaques and NFTs. Aggregated Aß can activate complement, leading to synapse pruning and loss by microglial phagocytosis. Systemic inflammation can activate microglial TLR4, NLRP3 inflammasome, and complement in the brain, leading to neuroinflammation, Aß accumulation, synapse loss and neurodegeneration. The host immune response has been shown to function through complex crosstalk between the TLR, complement and inflammasome signaling pathways. Accordingly, targeting the molecular mechanisms underlying the TLR-complement-NLRP3 inflammasome signaling pathways can be a preventive and therapeutic approach for AD.


Assuntos
Doença de Alzheimer/imunologia , Proteínas do Sistema Complemento/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Camundongos
13.
J Neuroinflammation ; 5: 23, 2008 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-18510752

RESUMO

BACKGROUND: Abeta deposits in the brains of patients with Alzheimer's disease (AD) are closely associated with innate immune responses such as activated microglia and increased cytokines. Accumulating evidence supports the hypothesis that innate immune/inflammatory responses play a pivotal role in the pathogenesis of AD: either beneficial or harmful effects on the AD progression. The molecular mechanisms by which the innate immune system modulates the AD progression are not well understood. Toll-like receptors (TLRs) are first-line molecules for initiating the innate immune responses. When activated through TLR signaling, microglia respond to pathogens and damaged host cells by secreting chemokines and cytokines and express the co-stimulatory molecules needed for protective immune responses to pathogens and efficient clearance of damaged tissues. We previously demonstrated that an AD mouse model homozygous for a destructive mutation of TLR4 has increases in diffuse and fibrillar Abeta deposits as well as buffer-soluble and insoluble Abeta in the brain as compared with a TLR4 wild-type AD mouse model. Here, we investigated the roles of TLR4 in Abeta-induced upregulation of cytokines and chemokines, Abeta-induced activation of microglia and astrocytes and Abeta-induced immigration of leukocytes. METHODS: Using the same model, levels of cytokines and chemokines in the brain were determined by multiplex cytokine/chemokine array. Activation of microglia and astrocytes and immigration of leukocytes were determined by immunoblotting and immunohistochemistry followed by densitometry and morphometry, respectively. RESULTS: Levels of tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, IL-10 and IL-17 in the brains of TLR4 wild-type AD mice were significantly higher than those in TLR4 wild-type non-transgenic littermates. Such increases in cytokines were not found in TLR4 mutant AD mice as compared with TLR4 mutant non-transgenic littermates. Although expression levels of CD11b (a microglia marker) and GFAP (a reactive astrocyte marker) in the brains of TLR4 mutant AD mice were higher than those in TLR4 wild type AD mice, no difference was found in levels of CD45 (common leukocyte antigen). CONCLUSION: This is the first demonstration of TLR4-dependent upregulation of cytokines in an AD mouse model. Our results suggest that TLR4 signaling is involved in AD progression and that TLR4 signaling can be a new therapeutic target for AD.


Assuntos
Doença de Alzheimer/metabolismo , Citocinas/biossíntese , Receptor 4 Toll-Like/fisiologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Astrócitos/metabolismo , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Quimiotaxia de Leucócito , Citocinas/genética , Feminino , Regulação da Expressão Gênica , Humanos , Interleucina-10/biossíntese , Interleucina-10/genética , Interleucina-17/biossíntese , Interleucina-17/genética , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Mutação Puntual , Presenilina-1/genética , Proteínas Recombinantes de Fusão/genética , Deleção de Sequência , Organismos Livres de Patógenos Específicos , Receptor 4 Toll-Like/deficiência , Receptor 4 Toll-Like/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Regulação para Cima
14.
Immunol Lett ; 112(1): 30-8, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17686533

RESUMO

Accumulation of aggregated amyloid beta-protein (Abeta) in the brain is thought to be the initiating event leading to neurodegeneration and dementia in Alzheimer's disease (AD). Therefore, therapeutic strategies that clear accumulated Abeta and/or prevent Abeta production and its aggregation are predicted to be effective against AD. Immunization of AD mouse models with synthetic Abeta prevented or reduced Abeta load in the brain and ameliorated their memory and learning deficits. The clinical trials of Abeta immunization elicited immune responses in only 20% of AD patients and caused T-lymphocyte meningoencephalitis in 6% of AD patients. In attempting to develop safer vaccines, we previously demonstrated that an adenovirus vector, AdPEDI-(Abeta1-6)11, which encodes 11 tandem repeats of Abeta1-6 can induce anti-inflammatory Th2 immune responses in mice. Here, we investigated whether a DNA prime-adenovirus boost regimen could elicit a more robust Th2 response using AdPEDI-(Abeta1-6)11 and a DNA plasmid encoding the same antigen. All mice (n=7) subjected to the DNA prime-adenovirus boost regimen were positive for anti-Abeta antibody, while, out of 7 mice immunized with only AdPEDI-(Abeta1-6)11, four mice developed anti-Abeta antibody. Anti-Abeta titers were indiscernible in mice (n=7) vaccinated with only DNA plasmid. The mean anti-Abeta titer induced by the DNA prime-adenovirus boost regimen was approximately 7-fold greater than that by AdPEDI-(Abeta1-6)11 alone. Furthermore, anti-Abeta antibodies induced by the DNA prime-adenovirus boost regimen were predominantly of the IgG1 isotype. These results indicate that the DNA prime-adenovirus boost regimen can enhance Th2-biased responses with AdPEDI-(Abeta1-6)11 in mice and suggest that heterologous prime-boost strategies may make AD immunotherapy more effective in reducing accumulated Abeta.


Assuntos
Adenoviridae/genética , Vacinas contra Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Vetores Genéticos , Imunização Secundária , Fragmentos de Peptídeos/imunologia , Células Th2/imunologia , Vacinas de DNA/imunologia , Administração Intranasal , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Vacinas contra Alzheimer/administração & dosagem , Vacinas contra Alzheimer/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Formação de Anticorpos , Encéfalo/imunologia , Encéfalo/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina G/sangue , Interferon gama/sangue , Interleucina-10/sangue , Interleucina-2/sangue , Interleucina-4/sangue , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fatores de Tempo , Transfecção , Vacinas de DNA/administração & dosagem , Vacinas de DNA/metabolismo
15.
Brain ; 129(Pt 11): 3006-19, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16984903

RESUMO

Deposits of amyloid beta-protein (Abeta) in neuritic plaques and cerebral vessels are a pathological hallmark of Alzheimer's disease. Fibrillar Abeta deposits are closely associated with inflammatory responses such as activated microglia in brain with this disease. Increasing lines of evidence support the hypothesis that activated microglia, innate immune cells in the CNS, play a pivotal role in the progression of the disease: either clearing Abeta deposits by phagocytic activity or releasing cytotoxic substances and pro-inflammatory cytokines. Toll-like receptors (TLRs) are a family of pattern-recognition receptors in the innate immune system. Exogenous and endogenous TLR ligands activate microglia. To investigate the role of TLR4 in the amyloidogenesis in vivo, we determined the amounts of cerebral Abeta in Alzheimer's disease mouse models with different genotypes of TLR4 using three distinct methods. We show that mouse models (Mo/Hu APPswe PS1dE9 mice) homozygous for a destructive mutation of TLR4 (Tlr(Lps-d)/Tlr(Lps-d)) had increases in diffuse and fibrillar Abeta deposits by immunocytochemistry, fibrillar Abeta deposits by thioflavine-S staining and buffer-soluble and insoluble Abeta by ELISA in the cerebrum, as compared with TLR4 wild-type mouse models. Although the differences in these parameters were less significant, mouse models heterozygous for the mutation (Tlr(Lps-d)/) showed co-dominant phenotypes. Consistent with these observations in vivo, cultured microglia derived from Tlr(Lps-d)/Tlr(Lps-d) mice failed to show an increase in Abeta uptake after stimulation with a TLR4 ligand but not with a TLR9 ligand in vitro. Furthermore, activation of microglia (BV-2 cell) with a TLR2, TLR4 or TLR9 ligand, markedly boosted ingestion of Abeta in vitro. These results suggest that TLR signalling pathway(s) may be involved in clearance of Abeta-deposits in the brain and that TLRs can be a therapeutic target for Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Receptor 4 Toll-Like/fisiologia , Doença de Alzheimer/genética , Animais , Western Blotting/métodos , Encéfalo/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Genótipo , Ligantes , Camundongos , Microglia/metabolismo , Mutação , Fragmentos de Peptídeos/metabolismo , Presenilina-1/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor Toll-Like 9/fisiologia
16.
Mol Neurobiol ; 51(1): 43-56, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24733587

RESUMO

Accumulation of amyloid beta-peptide (Aß) in the brain is hypothesized to be a causal event leading to dementia in Alzheimer's disease (AD). Aß vaccination removes Aß deposits from the brain. Aß immunotherapy, however, may cause T cell- and/or Fc-receptor-mediated brain inflammation and relocate parenchymal Aß deposits to blood vessels leading to cerebral hemorrhages. Because catalytic antibodies do not form stable immune complexes and Aß fragments produced by catalytic antibodies are less likely to form aggregates, Aß-specific catalytic antibodies may have safer therapeutic profiles than reversibly-binding anti-Aß antibodies. Additionally, catalytic antibodies may remove Aß more efficiently than binding antibodies because a single catalytic antibody can hydrolyze thousands of Aß molecules. We previously isolated Aß-specific catalytic antibody, IgVL5D3, with strong Aß-hydrolyzing activity. Here, we evaluated the prophylactic and therapeutic efficacy of brain-targeted IgVL5D3 gene delivery via recombinant adeno-associated virus serotype 9 (rAAV9) in an AD mouse model. One single injection of rAAV9-IgVL5D3 into the right ventricle of AD model mice yielded widespread, high expression of IgVL5D3 in the unilateral hemisphere. IgVL5D3 expression was readily detectable in the contralateral hemisphere but to a much lesser extent. IgVL5D3 expression was also confirmed in the cerebrospinal fluid. Prophylactic and therapeutic injection of rAAV9-IgVL5D3 reduced Aß load in the ipsilateral hippocampus of AD model mice. No evidence of hemorrhages, increased vascular amyloid deposits, increased proinflammatory cytokines, or infiltrating T-cells in the brains was found in the experimental animals. AAV9-mediated anti-Aß catalytic antibody brain delivery can be prophylactic and therapeutic options for AD.


Assuntos
Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/terapia , Biocatálise , Genes de Imunoglobulinas , Terapia Genética , Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/complicações , Peptídeos beta-Amiloides/metabolismo , Animais , Anticorpos/farmacologia , Biocatálise/efeitos dos fármacos , Angiopatia Amiloide Cerebral/etiologia , Angiopatia Amiloide Cerebral/patologia , Hemorragia Cerebral/etiologia , Hemorragia Cerebral/patologia , Dependovirus/metabolismo , Modelos Animais de Doenças , Vetores Genéticos/metabolismo , Células HEK293 , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Inflamação/patologia , Injeções , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/patologia , Neocórtex/efeitos dos fármacos , Neocórtex/metabolismo , Neocórtex/patologia , Placa Amiloide/metabolismo , Solubilidade
17.
Neurosci Lett ; 370(2-3): 218-23, 2004 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-15488326

RESUMO

Induction of anti-amyloid beta-protein (Abeta) antibodies in transgenic mouse models of Alzheimer disease (AD) by repeated injection of synthetic Abeta was shown to be effective in preventing and removing deposition of Abeta aggregates in the brain. Here, we have tested a non-invasive modality whereby a replication-defective adenovirus vector encoding Abeta was intranasally administered to mice to elicit immune responses against Abeta. Intranasal immunization only with the adenovirus vector failed to induce significant immune responses. When an adenovirus vector encoding granulocyte/macrophage-colony stimulating factor (GM-CSF) was used as an adjuvant in conjunction with the adenovirus encoding Abeta, a marked immune response was elicited against Abeta. Immunoglobulin isotyping revealed that the induced anti-Abeta antibodies are predominantly of the IgG2b and IgG1 isotypes, suggesting a Th-2 anti-inflammatory type. Furthermore, amyloid load in the brain of AD model mice (Tg2576) vaccinated with adenovirus vectors encoding Abeta and GM-CSF was much smaller than that in control Tg2576 mice. Thus, intranasal administration of adenovirus vectors encoding Abeta and GM-CSF may be effective in prevention and treatment of AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Amiloide/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fragmentos de Peptídeos/imunologia , Adenoviridae/genética , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Terapia Genética , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Imunização , Imunoglobulinas/classificação , Imunoglobulinas/uso terapêutico , Imuno-Histoquímica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fragmentos de Peptídeos/genética , Fatores de Tempo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
18.
J Mol Neurosci ; 49(2): 277-88, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22945846

RESUMO

We previously reported that anti-amyloid-beta (Aß) single-chain antibody (scFv59) brain delivery via recombinant adeno-associated virus (rAAV) was effective in reducing cerebral Aß load in an Alzheimer's disease (AD) mouse model without inducing inflammation. Here, we investigated the prophylactic effects and mechanism of a muscle-directed gene therapy modality in an AD mouse model. We injected rAAV serotype 1 encoding scFv59 into the right thigh muscles of 3-month-old mice. Nine months later, high levels of scFv59 expression were confirmed in the thigh muscles by both immunoblotting and immunohistochemistry. As controls, model mice were similarly injected with rAAV1 encoding antihuman immunodeficiency virus Gag antibody (scFvGag). AAV1-mediated scFv59 gene delivery was effective in decreasing Aß deposits in the brain. Compared with the scFvGag group, levels of Aß in cerebrospinal fluid (CSF) decreased significantly while Aß in serum tended to increase in the scFv59 group. AAV1-mediated scFv59 gene delivery may alter the equilibrium of Aß between the blood and brain, resulting in an increased efflux of Aß from the brain owing to antibody-mediated sequestration/clearance of peripheral Aß. Our results suggest that muscle-directed scFv59 delivery via rAAV1 may be a prophylactic option for AD and that levels of CSF Aß may be used to evaluate the efficacy of anti-Aß immunotherapy.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Anticorpos de Cadeia Única/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/sangue , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Peptídeos beta-Amiloides/imunologia , Animais , Dependovirus/genética , Modelos Animais de Doenças , Expressão Gênica , Marcação de Genes , Terapia Genética , Imunização Passiva , Camundongos , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Anticorpos de Cadeia Única/metabolismo
19.
J Neuroimmunol ; 244(1-2): 70-83, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22326143

RESUMO

We evaluated the therapeutic efficacy of combined treatment of Aß-immunization with simvastatin in an Alzheimer mouse model at age 22 months. DNA prime-adenovirus boost immunization induced modest anti-Aß titers and simvastatin increased the seropositive rate. Aß-KLH was additionally administered to boost the titers. Irrespective of simvastatin, the immunization did not decrease cerebral Aß deposits but increased soluble Aß and tended to exacerbate amyloid angiopathy in the hippocampus. The immunization increased cerebral invasion of leukocytes and simvastatin counteracted the increase. Thus, modest anti-Aß titers can increase soluble Aß and simvastatin may reduce inflammation associated with vaccination in aged Alzheimer mouse models.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Vacinas contra Alzheimer/imunologia , Peptídeos beta-Amiloides/imunologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Sinvastatina/uso terapêutico , Doença de Alzheimer/imunologia , Doença de Alzheimer/prevenção & controle , Vacinas contra Alzheimer/uso terapêutico , Animais , Angiopatia Amiloide Cerebral/tratamento farmacológico , Angiopatia Amiloide Cerebral/imunologia , Angiopatia Amiloide Cerebral/prevenção & controle , Modelos Animais de Doenças , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
20.
Behav Brain Res ; 227(1): 36-42, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22051943

RESUMO

Toll-like receptors (TLRs) are a family of pattern-recognition receptors in innate immunity and provide a first line defense against pathogens and tissue injuries. In addition to important roles in infection, inflammation, and immune diseases, recent studies show that TLR signaling is involved in modulation of learning, memory, mood, and neurogenesis. Because MyD88 is essential for the downstream signaling of all TLRs, except TLR3, we investigated the effects of MyD88 deficiency (MyD88-/-) on behavioral functions in mice. Additionally, we recently demonstrated that a mouse model of Alzheimer's disease (AD) deficient for MyD88 had decreases in Aß deposits and soluble Aß in the brain as compared with MyD88 sufficient AD mouse models. Because accumulation of Aß in the brain is postulated to be a causal event leading to cognitive deficits in AD, we investigated the effects of MyD88 deficiency on behavioral functions in the AD mouse model at 10 months of age. MyD88 deficient mice showed more anxiety in the elevated plus-maze. In the motor coordination tests, MyD88 deficient mice remained on a beam and a bar for a longer time, but with slower initial movement on the bar. In the Morris water maze test, MyD88 deficiency appeared to improve spatial learning irrespective of the transgene. Our findings suggest that the MyD88-dependent pathway contributes to behavioral functions in an AD mouse model and its control group.


Assuntos
Doença de Alzheimer/complicações , Ansiedade/etiologia , Comportamento Exploratório/fisiologia , Atividade Motora/fisiologia , Fator 88 de Diferenciação Mieloide/deficiência , Desempenho Psicomotor/fisiologia , Percepção Espacial/fisiologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Humanos , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Presenilina-1/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA