Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
2.
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
3.
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
4.
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
5.
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
6.
J Neuroimmunol ; 299: 98-106, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27725131

RESUMO

Naturally-occurring bioactive flavonoids such as diosmin significantly reduces amyloid beta (Aß) associated pathology in Alzheimer's disease (AD) mouse models. In the present study, oral administration of diosmin reduced cerebral Aß oligomer levels, tau-hyperphosphorylation and cognitive impairment in the 3xTg-AD mouse model through glycogen synthase kinase-3 (GSK-3) and transient receptor potential canonical 6-related mechanisms. Diosmetin, one major bioactive metabolite of diosmin, increased inhibitory GSK-3ß phosphorylation, while selectively reducing γ-secretase activity, Aß generation, tau hyperphosphorylation and pro-inflammatory activation of microglia in vitro, without altering Notch processing. Therefore, both diosmin and diosmetin could be considered as potential candidates for novel anti-AD therapy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Córtex Cerebral/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Diosmina/uso terapêutico , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Células CHO , Córtex Cerebral/efeitos dos fármacos , Disfunção Cognitiva/metabolismo , Cricetinae , Cricetulus , Diosmina/farmacologia , Relação Dose-Resposta a Droga , Feminino , Células HeLa , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas tau/antagonistas & inibidores
7.
Cell Transplant ; 25(11): 1967-1977, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-28836850

RESUMO

The prevalence of HIV-associated neurocognitive disorders (HAND) remains high despite combination antiretroviral therapy (cART). There is evidence that neural stem cells (NSCs) can migrate to sites of brain injury such as those caused by inflammation and oxidative stress, which are pathological features of HAND. Thus, reductions in NSCs may contribute to HAND pathogenesis. Since the HIV non-nucleoside reverse transcriptase inhibitor efavirenz (EFV) has previously been associated with cognitive deficits and promotion of oxidative stress pathways, we examined its effect on NSCs in vitro as well as in C57BL/6J mice. Here we report that EFV induced a decrease in NSC proliferation in vitro as indicated by MTT assay, as well as BrdU and nestin immunocytochemistry. In addition, EFV decreased intracellular NSC adenosine triphosphate (ATP) stores and NSC mitochondrial membrane potential (MMP). Further, we found that EFV promoted increased lactate dehydrogenase (LDH) release, activation of p38 mitogen-activated protein kinase (MAPK), and increased Bax expression in cultured NSCs. Moreover, EFV reduced the quantity of proliferating NSCs in the subventricular zone (SVZ) of C57BL/6J mice as suggested by BrdU, and increased apoptosis as measured by active caspase-3 immunohistochemistry. If these in vitro and in vivo models translate to the clinical syndrome, then a pharmacological or cell-based therapy aimed at opposing EFV-mediated reductions in NSC proliferation may be beneficial to prevent or treat HAND in patients receiving EFV.


Assuntos
Benzoxazinas/toxicidade , Proliferação de Células/efeitos dos fármacos , Inibidores da Transcriptase Reversa/toxicidade , Trifosfato de Adenosina/metabolismo , Alcinos , Animais , Apoptose/efeitos dos fármacos , Benzoxazinas/administração & dosagem , Caspase 3/metabolismo , Células Cultivadas , Ciclopropanos , Feminino , Imuno-Histoquímica , Injeções Intraperitoneais , Ventrículos Laterais/efeitos dos fármacos , Ventrículos Laterais/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Inibidores da Transcriptase Reversa/administração & dosagem , Proteína X Associada a bcl-2/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
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
9.
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
10.
Cell Transplant ; 24(11): 2237-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26230612

RESUMO

Alzheimer's disease (AD) is the fourth major cause of mortality in the elderly in the US and the leading cause of dementia worldwide. While pharmacological targets have been discovered, there are no true disease-modifying therapies. We have recently discovered that multiple low-dose infusions of human umbilical cord blood cells (HUCBCs) ameliorate cognitive impairments and reduce Aß-associated neuropathology in PSAPP transgenic mice. However, the mechanism for these effects of HUCBCs remains unclear. In the present study, we examined whether monocytes, as important components of HUCBCs, would have beneficial outcomes on the reduction of AD-like pathology and associated cognitive impairments in PSAPP transgenic AD model mice. PSAPP mice and their wild-type littermates were treated monthly with an infusion of peripheral human umbilical cord blood cell (HUCBC)-derived monocytes over a period of 2 and 4 months, followed by behavioral evaluations, biochemical, and histological analyses. The principal findings of the present study confirmed that monocytes derived from HUCBCs (CB-M) play a central role in HUCBC-mediated cognition-enhancing and Aß pathology-ameliorating activities. Most importantly, we found that compared with CB-M, aged monocytes showed an ineffective phagocytosis of Aß, while exogenous soluble amyloid precursor protein α (sAPPα) could reverse this deficiency. Pretreating monocytes with sAPPα upregulates Aß internalization. Our further studies suggested that sAPPα could form a heterodimer with Aßs, with the APP672-688 (Aß1-16) region being responsible for this effect. This in turn promoted binding of these heterodimers to monocyte scavenger receptors and thus promoted enhanced Aß clearance. In summary, our findings suggest an interesting hypothesis that peripheral monocytes contribute to Aß clearance through heterodimerization of sAPPα with Aß. Further, declined or impaired sAPPα production, or reduced heterodimerization with Aß, would cause a deficiency in Aß clearance and thus accelerate the pathogenesis of AD.


Assuntos
Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Monócitos/transplante , Cordão Umbilical/citologia , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Humanos , Camundongos , Monócitos/metabolismo , Domínios e Motivos de Interação entre Proteínas
11.
Sci Rep ; 5: 11322, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-26091071

RESUMO

BACE1 initiates amyloid-ß (Aß) generation and the resultant cerebral amyloidosis, as a characteristic of Alzheimer's disease (AD). Thus, inhibition of BACE1 has been the focus of a large body of research. The most recent clinical trials highlight the difficulty involved in this type of anti-AD therapy as evidenced by side effects likely due to the ubiquitous nature of BACE1, which cleaves multiple substrates. The human Swedish mutant form of amyloid protein precursor (APPswe) has been shown to possess a higher affinity for BACE1 compared to wild-type APP (APPwt). We pursued a new approach wherein harnessing this greater affinity to modulate BACE1 APP processing activity. We found that one peptide derived from APPswe, containing the ß-cleavage site, strongly inhibits BACE1 activity and thereby reduces Aß production. This peptide, termed APPswe BACE1 binding site peptide (APPsweBBP), was further conjugated to the fusion domain of the HIV-1 Tat protein (TAT) at the C-terminus to facilitate its biomembrane-penetrating activity. APPwt and APPswe over-expressing CHO cells treated with this TAT-conjugated peptide resulted in a marked reduction of Aß and a significant increase of soluble APPα. Intraperitoneal administration of this peptide to 5XFAD mice markedly reduced ß-amyloid deposits as well as improved hippocampal-dependent learning and memory.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Sítios de Ligação , Mutação , Peptídeos/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/química , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Comportamento Animal , Sítios de Ligação/genética , Barreira Hematoencefálica/metabolismo , Células CHO , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Cricetulus , Modelos Animais de Doenças , Expressão Gênica , Hipocampo/metabolismo , Memória , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Conformação Proteica , Proteólise , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
12.
Am J Transl Res ; 7(2): 328-38, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25893035

RESUMO

HIV-1 Tat protein is a key neuropathological element in HIV associated neurogcognitive disorders (HAND); a type of cognitive syndrome thought to be at least partially mediated by increased levels of brain reactive oxygen species (ROS) and nitric oxide (NO). Methylsulfonylmethane (MSM) is a sulfur-containing compound known to reduce oxidative stress. This study was conducted to determine whether administration of MSM attenuates HIV-1 Tat induced oxidative stress in mouse neuronal cells. MSM treatment significantly decreased neuronal cell NO and ROS secretion. Further, MSM significantly reversed HIV-1 Tat mediated reductions in reduced glutathione (GSH) as well as HIV-1 Tat mediated increases in oxidized glutathione (GSSG). In addition, Tat reduced nuclear translocation of nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), a key nuclear promoter of antioxidant activity, while MSM increased its translocation to the nucleus in the presence of Tat. These results suggest that HIV-1 Tat reduces the resiliency of neuron cells to oxidative stress which can be reversed by MSM. Given the clinical safety of MSM, future preclinical in vivo studies will be required to further confirm these results in effort to validate MSM as a neuroprotectant in patients at risk of, or who are already diagnosed with, HAND.

13.
Mol Neurobiol ; 52(1): 115-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25119777

RESUMO

Amyloid-beta (Aß) plays a central role in the pathogenesis of Alzheimer's disease (AD), and it is a major therapeutic target for AD. It is proposed that removal of Aß in blood can facilitate Aß clearance from the brain, representing a promising therapeutic approach for AD. However, the efficacy and mechanisms for Aß clearance by peripheral organs and tissues remain largely unknown. In the present study, 47 chronic kidney disease (CKD) patients (16 newly diagnosed patients who had never been dialyzed and 31 patients who were receiving dialysis) and 43 normal controls (NC) were enrolled. We found that serum Aß levels were significantly higher in CKD patients than NC. CKD patients who were receiving dialysis had lower serum Aß levels than patients without receiving dialysis, being comparable to NC. Furthermore, serum Aß levels were correlated with renal functions reflected by estimated glomerular filtration rate (eGFR) and residual GFR (rGFR). Our study suggests that kidney is involved in peripheral clearance of Aß, and dialysis might be a potential therapeutic approach of Aß removal.


Assuntos
Peptídeos beta-Amiloides/sangue , Rim/metabolismo , Demografia , Feminino , Humanos , Rim/fisiopatologia , Masculino , Pessoa de Meia-Idade , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/fisiopatologia
14.
Mol Neurobiol ; 51(1): 1-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24733588

RESUMO

Amyloid-beta (Aß) is suggested to play a causal role in the pathogenesis of Alzheimer's disease (AD). Immunotherapies are among the most promising Aß-targeting therapeutic strategies for AD. But, to date, all clinical trials of this modality have not been successful including Aß vaccination (AN1792), anti-Aß antibodies (bapineuzumab, solanezumab and ponezumab), and intravenous immunoglobulin (IVIG). We propose that one reason for the failures of these clinical trials may be the adverse effects of targeting the central clearance of amyloid plaques. The potential adverse effects include enhanced neurotoxicity related to Aß oligomerization from plaques, neuroinflammation related to opsonized Aß phagocytosis, autoimmunity related to cross-binding of antibodies to amyloid precursor protein (APP) on the neuron membrane, and antibody-mediated vascular and neuroskeletal damage. Overall, the majority of the adverse effects seen in clinical trials were associated with the entry of antibodies into the brain. Finally, we propose that peripheral Aß clearance would be effective and safe for future Aß-targeting therapies.


Assuntos
Doença de Alzheimer/sangue , Peptídeos beta-Amiloides/sangue , Encéfalo/patologia , Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Encéfalo/metabolismo , Humanos , Imunoterapia , Modelos Biológicos
15.
Mol Neurodegener ; 9: 40, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25304757

RESUMO

Given the increased life expectancy of human immunodeficiency virus (HIV) infected individuals treated with combination antiretroviral therapy (cART) and the ongoing inflammation observed in the brains of these patients, it is likely that premature neurodegeneration as measured by phospho-tau (p-tau) or increased total tau (t-tau) protein may become an increasing problem. This review examines the seven human studies that have occurred over the past 14 years measuring p-tau and/or t-tau in cerebrospinal fluid (CSF) or via post-mortem brain immunohistochemistry. Although not all studies are in agreement as to the changes in p-and t-tau in HIV infected patients, HIV persists in the brain despite cART. Thus is it is suggested that those maintained on long-term cART may develop tau pathology beyond the extent seen in the studies reviewed herein and overtime may then reach the threshold for clinical manifestation.


Assuntos
Encéfalo/patologia , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/patologia , Proteínas tau/metabolismo , Encéfalo/metabolismo , Humanos
16.
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
17.
PLoS One ; 9(4): e95500, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24759994

RESUMO

Efavirenz (EFV) is among the most commonly used antiretroviral drugs globally, causes neurological symptoms that interfere with adherence and reduce tolerability, and may have central nervous system (CNS) effects that contribute in part to HIV associated neurocognitive disorders (HAND) in patients on combination antiretroviral therapy (cART). Thus we evaluated a commonly used EFV containing regimen: EFV/zidovudine (AZT)/lamivudine (3TC) in murine N2a cells transfected with the human "Swedish" mutant form of amyloid precursor protein (SweAPP N2a cells) to assess for promotion of amyloid-beta (Aß) production. Treatment with EFV or the EFV containing regimen generated significantly increased soluble amyloid beta (Aß), and promoted increased ß-secretase-1 (BACE-1) expression while 3TC, AZT, or, vehicle control did not significantly alter these endpoints. Further, EFV or the EFV containing regimen promoted significantly more mitochondrial stress in SweAPP N2a cells as compared to 3TC, AZT, or vehicle control. We next tested the EFV containing regimen in Aß - producing Tg2576 mice combined or singly using clinically relevant doses. EFV or the EFV containing regimen promoted significantly more BACE-1 expression and soluble Aß generation while 3TC, AZT, or vehicle control did not. Finally, microglial Aß phagocytosis was significantly reduced by EFV or the EFV containing regimen but not by AZT, 3TC, or vehicle control alone. These data suggest the majority of Aß promoting effects of this cART regimen are dependent upon EFV as it promotes both increased production, and decreased clearance of Aß peptide.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Benzoxazinas/uso terapêutico , Alcinos , Animais , Benzoxazinas/administração & dosagem , Linhagem Celular , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Ciclopropanos , Feminino , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Lamivudina/administração & dosagem , Lamivudina/uso terapêutico , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Inibidores da Transcriptase Reversa/administração & dosagem , Inibidores da Transcriptase Reversa/uso terapêutico , Zidovudina/administração & dosagem , Zidovudina/uso terapêutico
18.
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
19.
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
20.
Am J Transl Res ; 5(6): 634-42, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24093060

RESUMO

Accumulation of amyloid-ß (Aß) peptides (predominantly Aß40, 42) and their aggregation into plaques in the brain are thought to be the one of the major causes of Alzheimer's disease (AD). Originally discovered in our Chinese hamster ovary (CHO) cell line stably expressing human wild-type amyloid precursor protein (APP) (CHO/APPwt) cultures devoid of Aß production, we found that Mycoplasma selectively degrades soluble Aß in a time and dose (colony forming unit) dependent manner. Moreover, we fully characterized the Mycoplasma species as Mycoplasma hyorhinis (M. hyorhinis) by genetic and colony morphological analyses by light microscopy. Most interestingly, we attenuated the pathogenicity of M. hyorhinis by γ irradiation (3.5 Gy), and found that its ability to degrade Aß was retained. On the other hand, heated and sonicated M. hyorhinis failed to retain this ability to degrade Aß, suggesting that this degradation requires viable cells and likely a biologically active signaling pathway. In addition, we found that M. hyorhinis can degrade Aß produced in AD model mice (PSAPP mice) ex vivo. Finally, we found that irradiated (non-pathogenic) M. hyorhinis also can degrade Aß produced in PSAPP mice in vivo. These studies suggest that irradiated (non-pathogenic) M. hyorhinis can be a novel and alternative biological strategy for AD treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...