Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurochem ; 135(3): 630-7, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26342176

RESUMO

We recently found that sAPPα decreases amyloid-beta generation by directly associating with ß-site amyloid precursor protein (APP)-converting enzyme 1 (BACE1), thereby modulating APP processing. Because inhibition of BACE1 decreases glycogen synthase kinase 3 beta (GSK3ß)-mediated Alzheimer's disease (AD)-like tau phosphorylation in AD patient-derived neurons, we determined whether sAPPα also reduces GSK3ß-mediated tau phosphorylation. We initially found increased levels of inhibitory phosphorylation of GSK3ß (Ser9) in primary neurons from sAPPα over-expressing mice. Further, recombinant human sAPPα evoked the same phenomenon in SH-SY5Y cells. Further, in SH-SY5Y cells over-expressing BACE1, and HeLa cells over-expressing human tau, sAPPα reduced GSK3ß activity and tau phosphorylation. Importantly, the reductions in GSK3ß activity and tau phosphorylation elicited by sAPPα were prevented by BACE1 but not γ-secretase inhibition. In accord, AD mice over-expressing human sAPPα had less GSK3ß activity and tau phosphorylation compared with controls. These results implicate a direct relationship between APP ß-processing and GSK3ß-mediated tau phosphorylation and further define the central role of sAPPα in APP autoregulation and AD pathogenesis.


Assuntos
Precursor de Proteína beta-Amiloide/farmacologia , Quinase 3 da Glicogênio Sintase/fisiologia , Transdução de Sinais/fisiologia , Proteínas tau/antagonistas & inibidores , Proteínas tau/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Relação Dose-Resposta a Droga , Glicogênio Sintase Quinase 3 beta , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Transdução de Sinais/efeitos dos fármacos
2.
Acta Neuropathol ; 130(4): 487-99, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26363791

RESUMO

Amyloid-beta (Aß) plays a pivotal role in the pathogenesis of Alzheimer's disease (AD). The physiological capacity of peripheral tissues and organs in clearing brain-derived Aß and its therapeutic potential for AD remains largely unknown. Here, we measured blood Aß levels in different locations of the circulation in humans and mice, and used a parabiosis model to investigate the effect of peripheral Aß catabolism on AD pathogenesis. We found that blood Aß levels in the inferior/posterior vena cava were lower than that in the superior vena cava in both humans and mice. In addition, injected (125)I labeled Aß40 was located mostly in the liver, kidney, gastrointestinal tract, and skin but very little in the brain; suggesting that Aß derived from the brain can be cleared in the periphery. Parabiosis before and after Aß deposition in the brain significantly reduced brain Aß burden without alterations in the expression of amyloid precursor protein, Aß generating and degrading enzymes, Aß transport receptors, and AD-type pathologies including hyperphosphorylated tau, neuroinflammation, as well as neuronal degeneration and loss in the brains of parabiotic AD mice. Our study revealed that the peripheral system is potent in clearing brain Aß and preventing AD pathogenesis. The present work suggests that peripheral Aß clearance is a valid therapeutic approach for AD, and implies that deficits in the Aß clearance in the periphery might also contribute to AD pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Fragmentos de Peptídeos/metabolismo , Adulto , Idoso , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Radioisótopos do Iodo , Masculino , Camundongos Transgênicos , Pessoa de Meia-Idade , Presenilina-1/genética , Presenilina-1/metabolismo , Adulto Jovem
3.
Cell Mol Life Sci ; 71(22): 4387-92, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25134912

RESUMO

HIV-1 infection, in addition to its destructive effects on the immune system, plays a role in the development of neurocognitive deficits. Indeed up to 50% of long-term HIV infected patients suffer from HIV-associated neurocognitive disorders (HAND). These deficits have been well characterized and defined clinically according to a number of cognitive parameters. HAND is often accompanied by atrophy of the brain including inhibition of neurogenesis, especially in the hippocampus. Many mechanisms have been proposed as contributing factors to HAND including induction of oxidative stress in the central nervous system (CNS), chronic microglial-mediated neuroinflammation, amyloid-beta (Aß) deposition, hyperphosphorylated tau protein, and toxic effects of combination antiretroviral therapy (cART). In these review we focus solely on recent experimental evidence suggesting that disturbance by HIV-1 results in impairment of neurogenesis as one contributing factor to HAND. Impaired neurogenesis has been linked to cognitive deficits and other neurodegenerative disorders. This article will highlight recently identified pathological mechanisms which potentially contribute to the development of impaired neurogenesis by HIV-1 or HIV-1-associated proteins from both animal and human studies.


Assuntos
Infecções por HIV/complicações , Infecções por HIV/fisiopatologia , HIV-1/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurogênese , Peptídeos beta-Amiloides/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Humanos , Doenças Neurodegenerativas/etiologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
4.
Int J Mol Sci ; 15(1): 895-904, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24413756

RESUMO

Traumatic brain injury (TBI) occurs in response to an acute insult to the head and is recognized as a major risk factor for Alzheimer's disease (AD). Indeed, recent studies have suggested a pathological overlap between TBI and AD, with both conditions exhibiting amyloid-beta (Aß) deposits, tauopathy, and neuroinflammation. Additional studies involving animal models of AD indicate that some AD-related genotypic determinants may be critical factors enhancing temporal and phenotypic symptoms of TBI. Thus in the present study, we examined sub-acute effects of moderate TBI delivered by a gas-driven shock tube device in Aß depositing Tg2576 mice. Three days later, significant increases in b-amyloid deposition, glycogen synthase-3 (GSK-3) activation, phospho-tau, and pro-inflammatory cytokines were observed. Importantly, peripheral treatment with the naturally occurring flavonoid, luteolin, significantly abolished these accelerated pathologies. This study lays the groundwork for a safe and natural compound that could prevent or treat TBI with minimal or no deleterious side effects in combat personnel and others at risk or who have experienced TBI.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Lesões Encefálicas/complicações , Encéfalo/efeitos dos fármacos , Luteolina/uso terapêutico , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/metabolismo , Lesões Encefálicas/tratamento farmacológico , Citocinas/genética , Citocinas/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Luteolina/farmacocinética , Luteolina/farmacologia , Camundongos , Distribuição Tecidual , Proteínas tau/metabolismo
5.
J Neurosci Res ; 91(9): 1239-46, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23686791

RESUMO

Baicalein, a flavonoid isolated from the roots of Scutellaria baicalensis, is known to modulate γ-aminobutyric acid (GABA) type A receptors. Given prior reports demonstrating benefits of GABAA modulation for Alzheimer's disease (AD) treatment, we wished to determine whether this agent might be beneficial for AD. CHO cells engineered to overexpress wild-type amyloid precursor protein (APP), primary culture neuronal cells from AD mice (Tg2576) and AD mice were treated with baicalein. In the cell cultures, baicalein significantly reduced the production of ß-amyloid (Aß) by increasing APP α-processing. These effects were blocked by the GABAA antagonist bicuculline. Likewise, AD mice treated daily with i.p. baicalein for 8 weeks showed enhanced APP α-secretase processing, reduced Aß production, and reduced AD-like pathology together with improved cognitive performance. Our findings suggest that baicalein promotes nonamyloidogenic processing of APP, thereby reducing Aß production and improving cognitive performance, by activating GABAA receptors.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Antioxidantes/uso terapêutico , Flavanonas/uso terapêutico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Bicuculina/farmacologia , Células CHO , Cricetulus , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Antagonistas de Receptores de GABA-A/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Mutação/genética , Transfecção
6.
Mol Pharm ; 10(12): 4728-38, 2013 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-24191685

RESUMO

Current United States Food and Drug Administration (FDA)-approved lithium salts are plagued with a narrow therapeutic window. Recent attempts to find alternative drugs have identified new chemical entities, but lithium's polypharmacological mechanisms for treating neuropsychiatric disorders are highly debated and are not yet matched. Thus, re-engineering current lithium solid forms in order to optimize performance represents a low cost and low risk approach to the desired therapeutic outcome. In this contribution, we employed a crystal engineering strategy to synthesize the first ionic cocrystals (ICCs) of lithium salts with organic anions. We are unaware of any previous studies that have assessed the biological efficacy of any ICCs, and encouragingly we found that the new speciation did not negatively affect established bioactivities of lithium. We also observed that lithium ICCs exhibit modulated pharmacokinetics compared to lithium carbonate. Indeed, the studies detailed herein represent an important advancement in a crystal engineering approach to a new generation of lithium therapeutics.


Assuntos
Íons/química , Íons/farmacologia , Lítio/química , Lítio/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Íons/farmacocinética , Lítio/farmacocinética , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Óxido Nítrico/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Tecnologia Farmacêutica/métodos
7.
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
8.
J Neurochem ; 120(5): 732-40, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22188568

RESUMO

Several prior investigations of Alzheimer's disease (AD) patients have indicated naturally occurring autoantibodies against amyloid-ß (Aß) species are produced. Although many studies have focused on the relative concentrations or binding affinities of autoantibodies against Aß-related proteins in AD and aging, data regarding their functional properties are limited. It is generally believed that these antibodies act to aid in clearance of Aß. However, as antibodies which bind to Aß also typically bind to the parent amyloid precursor protein (APP), we reasoned that certain Aß-targeting autoantibodies may bind to APP thereby altering its conformation and processing. Here we show for the first time, that naturally occurring Aß-reactive autoantibodies isolated from AD patients, but not from healthy controls, promote ß-secretase activity in cultured cells. Furthermore, using monoclonal antibodies to various regions of Aß, we found that antibodies generated against the N-terminal region, especially Aß(1-17) , dose dependently promoted amyloidogenic processing of APP viaß-secretase activation. Thus, this property of certain autoantibodies in driving Aß generation could be of etiological importance in the development of sporadic forms of AD. Furthermore, future passive or active anti-Aß immunotherapies must consider potential off-target effects resulting from antibodies targeting the N-terminus of Aß, as co-binding to the corresponding region of APP may actually enhance Aß generation.


Assuntos
Doença de Alzheimer/imunologia , Secretases da Proteína Precursora do Amiloide/efeitos dos fármacos , Peptídeos beta-Amiloides/imunologia , Precursor de Proteína beta-Amiloide/metabolismo , Autoanticorpos/farmacologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/sangue , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Linhagem Celular , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Injeções Intraventriculares , Masculino , Camundongos , Pessoa de Meia-Idade
9.
J Neuroinflammation ; 9: 185, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22849382

RESUMO

A global health problem, traumatic brain injury (TBI) is especially prevalent in the current era of ongoing world military conflicts. Its pathological hallmark is one or more primary injury foci, followed by a spread to initially normal brain areas via cascades of inflammatory cytokines and chemokines resulting in an amplification of the original tissue injury by microglia and other central nervous system immune cells. In some cases this may predispose individuals to later development of Alzheimer's disease (AD). The inflammatory-based progression of TBI has been shown to be active in humans for up to 17 years post TBI. Unfortunately, all neuroprotective drug trials have failed, and specific treatments remain less than efficacious. These poor results might be explained by too much of a scientific focus on neurons without addressing the functions of microglia in the brain, which are at the center of proinflammatory cytokine generation. To address this issue, we provide a survey of the TBI-related brain immunological mechanisms that may promote progression to AD. We discuss these immune and microglia-based inflammatory mechanisms involved in the progression of post-trauma brain damage to AD. Flavonoid-based strategies to oppose the antigen-presenting cell-like inflammatory phenotype of microglia will also be reviewed. The goal is to provide a rationale for investigations of inflammatory response following TBI which may represent a pathological link to AD. In the end, a better understanding of neuroinflammation could open therapeutic avenues for abrogation of secondary cell death and behavioral symptoms that may mediate the progression of TBI to later AD.


Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/prevenção & controle , Lesões Encefálicas/imunologia , Lesões Encefálicas/prevenção & controle , Encéfalo/imunologia , Progressão da Doença , Doença de Alzheimer/epidemiologia , Animais , Encéfalo/efeitos dos fármacos , Lesões Encefálicas/epidemiologia , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia
10.
J Cell Mol Med ; 15(2): 327-38, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19900216

RESUMO

Mutations in the presenilin-1 (PS1) gene are independent causes of familial Alzheimer's disease (AD). AD patients have dysregulated immunity, and PS1 mutant mice exhibit abnormal systemic immune responses. To test whether immune function abnormality caused by a mutant human PS1 gene (mhPS1) could modify AD-like pathology, we reconstituted immune systems of AD model mice carrying a mutant human amyloid precursor protein gene (mhAPP; Tg2576 mice) or both mhAPP and mhPS1 genes (PSAPP mice) with allogeneic bone marrow cells. Here, we report a marked reduction in amyloid-ß (Aß) levels, ß-amyloid plaques and brain inflammatory responses in PSAPP mice following strain-matched wild-type PS1 bone marrow reconstitution. These effects occurred with immune switching from pro-inflammatory T helper (Th) 1 to anti-inflammatory Th2 immune responses in the periphery and in the brain, which likely instructed microglia to phagocytose and clear Aß in an ex vivo assay. Conversely, Tg2576 mice displayed accelerated AD-like pathology when reconstituted with mhPS1 bone marrow. These data show that haematopoietic cells bearing the mhPS1 transgene exacerbate AD-like pathology, suggesting a novel therapeutic strategy for AD based on targeting PS1 in peripheral immune cells.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/imunologia , Presenilina-1/genética , Presenilina-1/metabolismo , Células Th1/imunologia , Células Th2/imunologia , Doença de Alzheimer/imunologia , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Antígeno CD11b/biossíntese , Citocinas/biossíntese , Camundongos , Camundongos Transgênicos , Proteínas Mutantes/metabolismo , Mutação , Placa Amiloide
11.
Biochem Biophys Res Commun ; 398(3): 337-41, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20542014

RESUMO

Rapamycin is a well known immunosuppressant drug for rejection prevention in organ transplantation. Numerous clinical trials using rapamycin analogs, involving both children and adults with various disorders are currently ongoing worldwide. Most recently, rapamycin gained much attention for what appears to be life-span extending properties when administered to mice. The risk for Alzheimer disease (AD) is strongly and positively correlated with advancing age and is characterized by deposition of beta-amyloid peptides (Abeta) as senile plaques in the brain. We report that rapamycin (2.5muM), significantly increases Abeta generation in murine neuron-like cells (N2a) transfected with the human "Swedish" mutant amyloid precursor protein (APP). In concert with these observations, we found rapamycin significantly decreases the neuroprotective amino-terminal APP (amyloid precursor protein) cleavage product, soluble APP-alpha (sAPP-alpha) while increasing production of the beta-carboxyl-terminal fragment of APP (beta-CTF). These cleavage events are associated with decreased activation of a disintegrin and metallopeptidase domain-10 (ADAM-10), an important candidate alpha-secretase which opposes Abeta generation. To validate these findings in vivo, we intraperitoneal (i.p.) injected Tg2576 Abeta-overproducing transgenic mice with rapamycin (3mg/kg/day) for 2weeks. We found increased Abeta levels associated with decreased sAPP-alpha at an average rapamycin plasma concentration of 169.7+/-23.5ng/mL by high performance liquid chromatography (HPLC). These data suggest that although rapamycin may increase the lifespan in some mouse models, it may not decrease the risk for age-associated neurodegenerative disorders such as AD.


Assuntos
Proteínas ADAM/antagonistas & inibidores , Envelhecimento/efeitos dos fármacos , Doença de Alzheimer/induzido quimicamente , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Imunossupressores/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Sirolimo/farmacologia , Proteína ADAM10 , Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Imunossupressores/efeitos adversos , Imunossupressores/sangue , Camundongos , Camundongos Transgênicos , Sirolimo/efeitos adversos , Sirolimo/sangue
12.
Biol Psychiatry ; 86(3): 208-220, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31208706

RESUMO

BACKGROUND: The ɛ4 isoform of apolipoprotein E (apoE4) is a major genetic risk factor for the development of sporadic Alzheimer's disease (AD), and its modification has been an intense focus for treatment of AD during recent years. METHODS: We investigated the binding of apoE, a peptide corresponding to its low-density lipoprotein receptor binding domain (amino acids 133-152; ApoEp), and modified ApoEp to amyloid precursor protein (APP) and their effects on amyloid-ß (Aß) production in cultured cells. Having discovered a peptide (6KApoEp) that blocks the interaction of apoE with N-terminal APP, we investigated the effects of this peptide and ApoEp on AD-like pathology and behavioral impairment in 3XTg-AD and 5XFAD transgenic mice. RESULTS: ApoE and ApoEp, but not truncated apoE lacking the low-density lipoprotein receptor binding domain, physically interacted with N-terminal APP and thereby mediated Aß production. Interestingly, the addition of 6 lysine residues to the N-terminus of ApoEp (6KApoEp) directly inhibited apoE binding to N-terminal APP and markedly limited apoE- and ApoEp-mediated Aß generation, presumably through decreasing APP cellular membrane trafficking and p44/42 mitogen-activated protein kinase phosphorylation. Moreover, while promoting apoE interaction with APP by ApoEp exacerbated Aß and tau brain pathologies in 3XTg-AD mice, disrupting this interaction by 6KApoEp ameliorated cerebral Aß and tau pathologies, neuronal apoptosis, synaptic loss, and hippocampal-dependent learning and memory impairment in 5XFAD mice without altering cholesterol, low-density lipoprotein receptor, and apoE expression levels. CONCLUSIONS: These data suggest that disrupting apoE interaction with N-terminal APP may be a novel disease-modifying therapeutic strategy for AD.


Assuntos
Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Apolipoproteínas E/antagonistas & inibidores , Apolipoproteínas E/metabolismo , Encéfalo/patologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Apolipoproteínas E/genética , Células CHO , Cognição/efeitos dos fármacos , Cricetulus , Modelos Animais de Doenças , Feminino , Humanos , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Transgênicos
13.
J Neuroinflammation ; 5: 51, 2008 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19014446

RESUMO

Recently, the term "inflammaging" was coined by Franceshci and colleagues to characterize a widely accepted paradigm that ageing is accompanied by a low-grade chronic up-regulation of certain pro-inflammatory responses. Inflammaging differs significantly from the traditional five cardinal features of acute inflammation in that it is characterized by a relative decline in adaptive immunity and T-helper 2 responses and is associated with increased innate immunity by cells of the mononuclear phagocyte lineage. While the over-active innate immunity characteristic of inflammaging may remain subclinical in many elderly individuals, a portion of individuals (postulated to have a "high responder inflammatory genotype") may shift from a state of "normal" or "subclinical" inflammaging to one or more of a number of age-associated diseases. We and others have found that IFN-gamma and other pro-inflammatory cytokines interact with processing and production of Abeta peptide, the pathological hallmark feature of Alzheimer's disease (AD), suggesting that inflammaging may be a "prodrome" to AD. Although conditions of enhanced innate immune response with overproduction of pro-inflammatory proteins are associated with both healthy aging and AD, it is suggested that those who age "well" demonstrate anti-inflammaging mechanisms and biomarkers that likely counteract the adverse immune response of inflammaging. Thus, opposing the features of inflammaging may prevent or treat the symptoms of AD. In this review, we fully characterize the aging immune system. In addition, we explain how three novel treatments, (1) human umbilical cord blood cells (HUCBC), (2) flavanoids, and (3) Abeta vaccination oppose the forces of inflammaging and AD-like pathology in various mouse models.


Assuntos
Envelhecimento , Doença de Alzheimer/imunologia , Doença de Alzheimer/fisiopatologia , Sistema Imunitário/fisiologia , Inflamação , Envelhecimento/imunologia , Envelhecimento/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/administração & dosagem , Peptídeos beta-Amiloides/imunologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Modelos Animais de Doenças , Sangue Fetal , Flavonoides/uso terapêutico , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Vacinação
14.
Stem Cells Dev ; 17(3): 423-39, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18366296

RESUMO

Modulation of immune/inflammatory responses by diverse strategies including amyloid-beta (Abeta) immunization, nonsteroidal anti-inflammatory drugs, and manipulation of microglial activation states has been shown to reduce Alzheimer's disease (AD)-like pathology and cognitive deficits in AD transgenic mouse models. Human umbilical cord blood cells (HUCBCs) have unique immunomodulatory potential. We wished to test whether these cells might alter AD-like pathology after infusion into the PSAPP mouse model of AD. Here, we report a marked reduction in Abeta levels/beta-amyloid plaques and associated astrocytosis following multiple low-dose infusions of HUCBCs. HUCBC infusions also reduced cerebral vascular Abeta deposits in the Tg2576 AD mouse model. Interestingly, these effects were associated with suppression of the CD40-CD40L interaction, as evidenced by decreased circulating and brain soluble CD40L (sCD40L), elevated systemic immunoglobulin M (IgM) levels, attenuated CD40L-induced inflammatory responses, and reduced surface expression of CD40 on microglia. Importantly, deficiency in CD40 abolishes the effect of HUCBCs on elevated plasma Abeta levels. Moreover, microglia isolated from HUCBC-infused PSAPP mice demonstrated increased phagocytosis of Abeta. Furthermore, sera from HUCBC-infused PSAPP mice significantly increased microglial phagocytosis of the Abeta1-42 peptide while inhibiting interferon-gammainduced microglial CD40 expression. Increased microglial phagocytic activity in this scenario was inhibited by addition of recombinant CD40L protein. These data suggest that HUCBC infusion mitigates AD-like pathology by disrupting CD40L activity.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Vasos Sanguíneos/patologia , Terapia Baseada em Transplante de Células e Tecidos , Sangue Fetal/citologia , Sangue Fetal/transplante , Peptídeos beta-Amiloides/sangue , Animais , Astrócitos/patologia , Encéfalo/patologia , Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Contagem de Células , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Microglia/patologia , Peptídeos/imunologia , Fagocitose , Placa Amiloide/patologia , Células Th2/imunologia
15.
Rejuvenation Res ; 11(5): 891-901, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18789000

RESUMO

Alzheimer's Disease (AD) is the most common age-related dementia, with a current prevalence in excess of five million individuals in the United States. The aggregation of amyloid-beta (A beta) into fibrillar amyloid plaques is a key pathological event in the development of the disease. Microglial proinflammatory activation is widely known to cause neuronal and synaptic damage that correlates with cognitive impairment in AD. However, current pharmacological attempts at reducing neuroinflammation mediated via microglial activation have been largely negative in terms of slowing AD progression. Previously, we have shown that microglia express proinflammatory cytokines and a reduced capacity to phagocytose A beta in the context of CD40, A beta peptides and/or lipopolysaccharide (LPS) stimulation, a phenomenon that can be opposed by attenuation of p44/42 mitogen-activated protein kinase (MAPK) signaling. Other groups have found that blueberry (BB) extract both inhibits phosphorylation of this MAPK module and also improves cognitive deficits in AD model mice. Given these considerations and the lack of reduced A beta quantities in behaviorally improved BB-fed mice, we wished to determine whether BB supplementation would alter the microglial proinflammatory activation state in response to A beta. We found that BB significantly enhances microglial clearance of A beta, inhibits aggregation of A beta(1-42), and suppresses microglial activation, all via suppression of the p44/42 MAPK module. Thus, these data may explain the previously observed behavioral recovery in PSAPP mice and suggest a means by which dietary supplementation could mitigate an undesirable microglial response toward fibrillar A beta.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Mirtilos Azuis (Planta) , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Fragmentos de Peptídeos/toxicidade , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Peptídeos beta-Amiloides/metabolismo , Animais , Células Cultivadas , Dieta , Inibidores Enzimáticos/farmacologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Camundongos , Microglia/imunologia , Fragmentos de Peptídeos/metabolismo , Fagocitose/efeitos dos fármacos , Extratos Vegetais/farmacologia
16.
Cell Transplant ; 27(3): 438-455, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29560732

RESUMO

Alzheimer's disease (AD) is an age-related disorder that affects cognition. Our previous studies showed that the neuroprotective fragment of amyloid procurer protein (APP) metabolite, soluble APPα (sAPPα), interferes with ß-site APP-cleaving enzyme 1 (BACE1, ß-secretase) cleavage and reduces amyloid-ß (Aß) generation. In an attempt to identify approaches to restore sAPPα levels, we found that human cord blood serum (CBS) significantly promotes sAPPα production compared with adult blood serum (ABS) and aged blood serum (AgBS) in Chinese hamster ovary cells stably expressing wild-type human APP. Interestingly, CBS selectively mediated the α-secretase cleavage of human neuron-specific recombinant APP695 in a cell-free system independent of tumor necrosis factor-α converting enzyme (TACE; a disintegrin and metalloproteinase domain-containing protein 17 [ADAM17]) and ADAM. Subsequently, using 3-step chromatographic separation techniques (i.e., diethylaminoethanol, size-exclusion, and ion-exchange chromatography), we purified and ultimately identified a CBS-specific fraction with enhanced α-secretase catalytic activity (termed αCBSF) and found that αCBSF has more than 3,000-fold increased α-secretase catalytic activity compared with the original pooled CBS. Furthermore, intracerebroventricular injection of αCBSF markedly increased cerebral sAPPα levels together with significant decreases in cerebral Aß production and abnormal tau (Thr231) phosphorylation compared with the AgBS fraction with enhanced α-secretase activity (AgBSF) treatment in triple transgenic Alzheimer's disease (3xTg-AD) mice. Moreover, AgBSF administered intraperitoneally to transgenic mice with five familial Alzheimer's disease mutations (5XFAD) via an osmotic mini pump for 6 weeks (wk) ameliorated ß-amyloid plaques and reversed cognitive impairment measures. Together, our results propose the necessity for further study aimed at identification and characterization of α-secretase in CBS for novel and effective AD therapy.


Assuntos
Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Cricetulus , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Camundongos , Fosforilação , Proteínas tau/metabolismo
17.
Crit Rev Neurobiol ; 19(2-3): 79-118, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-19409058

RESUMO

Hepatitis-C virus (HCV) has infected an estimated 130 million people worldwide, most of whom are chronically infected. Infection is marked by both treatment- and non-treatment-related psychiatric symptoms. Symptoms associated with antiretroviral therapy, interferon-alpha (IFN-alpha), include acute confusional states, delirium, depression, irritability, and even mania. These psychiatric symptoms are further complicated by the high rate of substance abuse and comorbid HIV infection inherent to this population. Even in the absence of IFN-alpha therapy, comorbid depression, cognitive decline, and especially fatigue are common in patients suffering HCV. These comorbidities have significant effects on both treatments and outcomes, and thus are reviewed herein.


Assuntos
Hepatite C/psicologia , Transtornos Mentais/epidemiologia , Transtornos Mentais/etiologia , Antivirais/efeitos adversos , Antivirais/uso terapêutico , Transtorno Bipolar/induzido quimicamente , Encéfalo/virologia , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/epidemiologia , Comorbidade , Delírio/induzido quimicamente , Depressão/induzido quimicamente , Depressão/epidemiologia , Fadiga/epidemiologia , Infecções por HIV/complicações , Infecções por HIV/epidemiologia , Hepacivirus , Hepatite C/complicações , Hepatite C/tratamento farmacológico , Hepatite C/epidemiologia , Humanos , Interferon-alfa/efeitos adversos , Interferon-alfa/uso terapêutico , Humor Irritável/efeitos dos fármacos , Transtornos Mentais/induzido quimicamente , Transtornos Relacionados ao Uso de Substâncias/epidemiologia
18.
Neural Plast ; 2007: 79102, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17641738

RESUMO

High concentrations of pituitary adenylate cyclase-activating polypeptide (PACAP) nerve fibers are present in the central nucleus of amygdala (CeA), a brain region implicated in the control of fear-related behavior. This study evaluated PACAPergic modulation of fear responses at the CeA in male Sprague-Dawley rats. PACAP (50-100 pmol) microinfusion via intra-CeA cannulae produced increases in immobility and time the rats spent withdrawn into a corner opposite to the electrified probe compared to controls in the shock-probe fear/defensive burying test. Shock-probe burying and exploration, numbers of shocks received, locomotion distance, and velocity were all reduced by intra-CeA PACAP injection. Further, intra-CeA PACAP effects were manifested only when the animals were challenged by shock, as intra-CeA PACAP injections did not cause significant changes in the behaviors of unshocked rats. Thus, intra-CeA administration of PACAP produces a distinct reorganization of stress-coping behaviors from active (burying) to passive modes, such as withdrawal and immobility. These findings are potentially significant toward enhancing our understanding of the involvement of PACAP and the CeA in the neural basis of fear and anxiety.


Assuntos
Adaptação Psicológica/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Medo/efeitos dos fármacos , Microinjeções , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/administração & dosagem , Adaptação Psicológica/fisiologia , Tonsila do Cerebelo/fisiologia , Animais , Eletrochoque/métodos , Medo/fisiologia , Masculino , Microinjeções/métodos , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Ratos , Ratos Sprague-Dawley
19.
Neuromolecular Med ; 19(2-3): 300-308, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28612181

RESUMO

Previous studies have demonstrated that the low-density lipoprotein receptor-related protein-1 (LRP1) plays conflicting roles in Alzheimer's disease (AD) pathogenesis, clearing ß-amyloid (Aß) from the brain while also enhancing APP endocytosis and resultant amyloidogenic processing. We have recently discovered that co-expression of mutant LRP1 C-terminal domain (LRP1-CT C4408R) with Swedish mutant amyloid precursor protein (APPswe) in Chinese hamster ovary (CHO) cells decreases Aß production, while also increasing sAPPα and APP α-C-terminal fragment (α-CTF), compared with CHO cells expressing APPswe alone. Surprisingly, the location of this mutation on LRP1 corresponded with the α-secretase cleavage site of APP. Further experimentation confirmed that in CHO cells expressing APPswe or wild-type APP (APPwt), co-expression of LRP1-CT C4408R decreases Aß and increases sAPPα and α-CTF compared with co-expression of wild-type LRP1-CT. In addition, LRP1-CT C4408R enhanced the unglycosylated form of LRP1-CT and reduced APP endocytosis as determined by flow cytometry. This finding identifies a point mutation in LRP1 which slows LRP1-CT-mediated APP endocytosis and amyloidogenic processing, while enhancing APP α-secretase cleavage, thus demonstrating a potential novel target for slowing AD pathogenesis.


Assuntos
Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Mutação de Sentido Incorreto , Mutação Puntual , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Sequência de Bases , Células CHO , Cricetinae , Cricetulus , Endocitose , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/fisiologia , Domínios Proteicos , Proteínas Recombinantes/metabolismo
20.
Cell Death Dis ; 8(6): e2880, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28617434

RESUMO

Lithium has been marketed in the United States of America since the 1970s as a treatment for bipolar disorder. More recently, studies have shown that lithium can improve cognitive decline associated with Alzheimer's disease (AD). However, the current United States Food and Drug Administration-approved lithium pharmaceutics (carbonate and citrate chemical forms) have a narrow therapeutic window and unstable pharmacokinetics that, without careful monitoring, can cause serious adverse effects. Here, we investigated the safety profile, pharmacokinetics, and therapeutic efficacy of LISPRO (ionic co-crystal of lithium salicylate and l-proline), lithium salicylate, and lithium carbonate (Li2CO3). We found that LISPRO (8-week oral treatment) reduces ß-amyloid plaques and phosphorylation of tau by reducing neuroinflammation and inactivating glycogen synthase kinase 3ß in transgenic Tg2576 mice. Specifically, cytokine profiles from the brain, plasma, and splenocytes suggested that 8-week oral treatment with LISPRO downregulates pro-inflammatory cytokines, upregulates anti-inflammatory cytokines, and suppresses renal cyclooxygenase 2 expression in transgenic Tg2576 mice. Pharmacokinetic studies indicated that LISPRO provides significantly higher brain lithium levels and more steady plasma lithium levels in both B6129SF2/J (2-week oral treatment) and transgenic Tg2576 (8-week oral treatment) mice compared with Li2CO3. Oral administration of LISPRO for 28 weeks significantly reduced ß-amyloid plaques and tau-phosphorylation. In addition, LISPRO significantly elevated pre-synaptic (synaptophysin) and post-synaptic protein (post synaptic density protein 95) expression in brains from transgenic 3XTg-AD mice. Taken together, our data suggest that LISPRO may be a superior form of lithium with improved safety and efficacy as a potential new disease modifying drug for AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Compostos de Lítio/administração & dosagem , Prolina/administração & dosagem , Administração Oral , Doença de Alzheimer/tratamento farmacológico , Animais , Autofagia , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HeLa , Humanos , Inflamação , Carbonato de Lítio/administração & dosagem , Carbonato de Lítio/sangue , Compostos de Lítio/sangue , Compostos de Lítio/química , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Fagocitose , Fosforilação , Prolina/sangue , Prolina/química , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA