Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Mol Biol Rep ; 49(12): 12007-12015, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36273336

RESUMO

BACKGROUND: Interferon-gamma (IFN-γ) is an immune-derived cytokines in the innate and adaptive immune responses, and functions as a major pro-inflammatory cytokine. IFNγ has previously been reported involving in the regulation of bone metabolism. However, contradictory results about the roles of IFN-γ in bone formation or bone resorption have been reported. It is possible that the functions of IFN-γ in bone formation is dose-dependent or time-dependent. In this study we examined the effect of IFN-γ on different stages of osteoblastogenesis and bone formation. MATERIALS AND METHODS: Cell proliferation, gene expression and protein levels of the critical effectors involving in different stages of differentiation were compared between differentiating preosteoblast MC3T3-E1 treated with or without IFN-γ at different stages. Cell proliferation were determined by MTT assay. Expression levels of osteoblast differentiation markers was performed by quantitative PCR assay. Also, western blot was conducted to investigate the protein levels in those effectors. CONCLUSION: IFN-γ regulates osteoblast and bone formation in a stage-dependent manner. IFN-γ did not alter and the expression of critical osteogenic transcription factors, such as Runx2 and Cbfb, suggesting that the differentiation was not disrupted by IFN-γ. The cell number and the levels of matrix proteins, including COL1A and BSP, at both early and late stage of osteoblastogenesis were downregulated by IFN-γ, indicating its negative regulating roles in early stages. In contrast, the mineralization protein ALP and OCN was upregulated at late stages. The results suggested that IFN-γ might act as a negative regulator in osteoblast differentiation and bone formation at early stages but switch into positive regulator at late stage. Our data revealed the complex features of the effects of IFN-γ on osteoblast differentiation. The detailed mechanisms of how IFN-γ influence on the bone formation and balance of bone remodeling will be further studied.


Assuntos
Reabsorção Óssea , Osteogênese , Humanos , Interferon gama/farmacologia , Interferon gama/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Osteoblastos , Reabsorção Óssea/metabolismo , Remodelação Óssea , Diferenciação Celular
2.
J Exp Med ; 219(12)2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36107206

RESUMO

TREM2 is exclusively expressed by microglia in the brain and is strongly linked to the risk for Alzheimer's disease (AD). As microglial responses modulated by TREM2 are central to AD pathogenesis, enhancing TREM2 signaling has been explored as an AD therapeutic strategy. However, the effective therapeutic window targeting TREM2 is unclear. Here, by using microglia-specific inducible mouse models overexpressing human wild-type TREM2 (TREM2-WT) or R47H risk variant (TREM2-R47H), we show that TREM2-WT expression reduces amyloid deposition and neuritic dystrophy only during the early amyloid seeding stage, whereas TREM2-R47H exacerbates amyloid burden during the middle amyloid rapid growth stage. Single-cell RNA sequencing reveals suppressed disease-associated microglia (DAM) signature and reduced DAM population upon TREM2-WT expression in the early stage, whereas upregulated antigen presentation pathway is detected with TREM2-R47H expression in the middle stage. Together, our findings highlight the dynamic effects of TREM2 in modulating AD pathogenesis and emphasize the beneficial effect of enhancing TREM2 function in the early stage of AD development.


Assuntos
Doença de Alzheimer , Amiloidose , Doença de Alzheimer/patologia , Amiloide/metabolismo , Amiloidose/patologia , Animais , Encéfalo/patologia , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Microglia/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
3.
Mol Neurodegener ; 17(1): 57, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-36056345

RESUMO

BACKGROUND: The aggregation and spread of α-synuclein (α-Syn) protein and related neuronal toxicity are the key pathological features of Parkinson's disease (PD) and Lewy body dementia (LBD). Studies have shown that pathological species of α-Syn and tau can spread in a prion-like manner between neurons, although these two proteins have distinct pathological roles and contribute to different neurodegenerative diseases. It is reported that the low-density lipoprotein receptor-related protein 1 (LRP1) regulates the spread of tau proteins; however, the molecular regulatory mechanisms of α-Syn uptake and spread, and whether it is also regulated by LRP1, remain poorly understood. METHODS: We established LRP1 knockout (LRP1-KO) human induced pluripotent stem cells (iPSCs) isogenic lines using a CRISPR/Cas9 strategy and generated iPSC-derived neurons (iPSNs) to test the role of LRP1 in α-Syn uptake. We treated the iPSNs with fluorescently labeled α-Syn protein and measured the internalization of α-Syn using flow cytometry. Three forms of α-Syn species were tested: monomers, oligomers, and pre-formed fibrils (PFFs). To examine whether the lysine residues of α-Syn are involved in LRP1-mediated uptake, we capped the amines of lysines on α-Syn with sulfo-NHS acetate and then measured the internalization. We also tested whether the N-terminus of α-Syn is critical for LRP1-mediated internalization. Lastly, we investigated the role of Lrp1 in regulating α-Syn spread with a neuronal Lrp1 conditional knockout (Lrp1-nKO) mouse model. We generated adeno-associated viruses (AAVs) that allowed for distinguishing the α-Syn expression versus spread and injected them into the hippocampus of six-month-old Lrp1-nKO mice and the littermate wild type (WT) controls. The spread of α-Syn was evaluated three months after the injection. RESULTS: We found that the uptake of both monomeric and oligomeric α-Syn was significantly reduced in iPSNs with LRP1-KO compared with the WT controls. The uptake of α-Syn PFFs was also inhibited in LRP1-KO iPSNs, albeit to a much lesser extent compared to α-Syn monomers and oligomers. The blocking of lysine residues on α-Syn effectively decreased the uptake of α-Syn in iPSNs and the N-terminus of α-Syn was critical for LRP1-mediated α-Syn uptake. Finally, in the Lrp1-nKO mice, the spread of α-Syn was significantly reduced compared with the WT littermates. CONCLUSIONS: We identified LRP1 as a key regulator of α-Syn neuronal uptake, as well as an important mediator of α-Syn spread in the brain. This study provides new knowledge on the physiological and pathological role of LRP1 in α-Syn trafficking and pathology, offering insight for the treatment of synucleinopathies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , alfa-Sinucleína/metabolismo , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactente , Camundongos , Doença de Parkinson/metabolismo , Sinapsinas , Proteínas tau/metabolismo
4.
Neurobiol Aging ; 115: 20-28, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35453035

RESUMO

APOE4 is the first identified genetic risk factor and remains as the strongest predictor for late-onset Alzheimer's disease (AD). Studies of AD patients, AD patient-specific induced pluripotent stem cell-derived neurons and cerebral organoids, and human apoE4-expressing and apoE-deficient mouse models clearly demonstrate that apoE4 provokes neuroinflammation, impairs cerebrovasculature, and exacerbates amyloid and tau pathologies. ApoE expression is greatly up-regulated in disease-associated microglia in mouse models of amyloidosis and in human microglia from AD brains. Importantly, genetic knock-down or depletion of apoE in mice greatly attenuates neuroinflammation and alleviates amyloid and tau pathologies. Similar beneficial effects can be achieved when apoE reduction is induced by the overexpression of apoE metabolic receptor LDLR. Toward therapeutic implications, administration of apoE antisense oligonucleotides or apoE siRNAs leads to significant pharmacologic effects, i.e., significant alleviation of AD pathologies in mouse models. Therefore, apoE reduction represents a promising therapeutic strategy for the treatment of AD patients carrying the APOE ε4 allele. In this review, we summarize evidence and rationale on why and how we target apoE4 reduction for AD therapy.


Assuntos
Doença de Alzheimer , Apolipoproteína E4 , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/terapia , Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/metabolismo , Encéfalo/metabolismo , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos
5.
Neurology ; 98(20): e2036-e2045, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35314499

RESUMO

BACKGROUND AND OBJECTIVES: To identify clinicopathologic factors contributing to mild cognitive impairment (MCI) reversion to normal cognition. METHODS: We analyzed 3 longitudinal cohorts in this study: the Mayo Clinic Study of Aging (MCSA), the Religious Orders Study and Memory and Aging Project (ROSMAP), and the National Alzheimer's Coordinating Center (NACC). Demographic characteristics and clinical outcomes were compared between patients with MCI with or without an experience of reversion to normal cognition (referred to as reverters and nonreverters, respectively). We also compared longitudinal changes in cortical thickness, glucose metabolism, and amyloid and tau load in a subcohort of reverters and nonreverters in MCSA with MRI or PET imaging information from multiple visits. RESULTS: We identified 164 (56.4%) individuals in MCSA, 508 (66.8%) individuals in ROSMAP, and 280 (34.1%) individuals in NACC who experienced MCI reversion to normal cognition. Cox proportional hazards regression models showed that MCI reverters had an increased chance of being cognitively normal at the last visit in MCSA (HR 3.31, 95% CI 2.14-5.12), ROSMAP (HR 3.72, 95% CI 2.50-5.56), and NACC (HR 9.29, 95% CI 6.45-13.40) and a reduced risk of progression to dementia (HR 0.12, 95% CI 0.05-0.29 in MCSA; HR 0.41, 95% CI 0.32-0.53 in ROSMAP; and HR 0.29, 95% CI 0.21-0.40 in NACC). Compared with MCI nonreverters, reverters had better-preserved cortical thickness (ß = 0.082, p <0.001) and glucose metabolism (ß = 0.119, p = 0.001) and lower levels of amyloid, albeit statistically nonsignificant (ß = -0.172, p = 0.090). However, no difference in tau load was found between reverters and nonreverters (ß = 0.073, p = 0.24). DISCUSSION: MCI reversion to normal cognition is likely attributed to better-preserved cortical structure and glucose metabolism.


Assuntos
Doença de Alzheimer , Amiloidose , Disfunção Cognitiva , Envelhecimento , Doença de Alzheimer/patologia , Amiloide , Cognição , Disfunção Cognitiva/psicologia , Progressão da Doença , Glucose , Humanos , Testes Neuropsicológicos
6.
J Pers Med ; 11(7)2021 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-34357123

RESUMO

Breast magnetic resonance imaging (MRI) is currently a widely used clinical examination tool. Recently, MR diffusion-related technologies, such as intravoxel incoherent motion diffusion weighted imaging (IVIM-DWI), have been extensively studied by breast cancer researchers and gradually adopted in clinical practice. In this study, we explored automatic tumor detection by IVIM-DWI. We considered the acquired IVIM-DWI data as a hyperspectral image cube and used a well-known hyperspectral subpixel target detection technique: constrained energy minimization (CEM). Two extended CEM methods-kernel CEM (K-CEM) and iterative CEM (I-CEM)-were employed to detect breast tumors. The K-means and fuzzy C-means clustering algorithms were also evaluated. The quantitative measurement results were compared to dynamic contrast-enhanced T1-MR imaging as ground truth. All four methods were successful in detecting tumors for all the patients studied. The clustering methods were found to be faster, but the CEM methods demonstrated better performance according to both the Dice and Jaccard metrics. These unsupervised tumor detection methods have the advantage of potentially eliminating operator variability. The quantitative results can be measured by using ADC, signal attenuation slope, D*, D, and PF parameters to classify tumors of mass, non-mass, cyst, and fibroadenoma types.

7.
Int J Med Sci ; 18(10): 2137-2145, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33859520

RESUMO

Previous studies demonstrated that resveratrol (RES) is able to enhance antioxidant, anti-inflammatory and insulin actions in humans. It is unclear whether RES can be used as ergogenic aids to enhance high-intensity cycling exercise performance and attenuate the high-intensity exercise-induced oxidative stress and inflammation. This study investigated the effect of RES supplementation on oxidative stress, inflammation, exercise-induced fatigue, and endurance performance. Eight male athletes participated in this single-blind crossover designed study and randomly instructed to receive four days of either oral RES (480 mg per day, totally 1920mg) or placebo supplementation. The cycling exercise challenge at 80% maximal oxygen consumption with 60 rpm was performed following 4 days of either RES or placebo supplementation. The total cycling performance time was recorded. In addition, blood samples were obtained to analyze the changes in blood glucose, plasma non-esterified fatty acid, serum lactate dehydrogenase, creatine kinase, uric acid, total antioxidant capacity, malondialdehyde, tumor necrosis factor-α, and interleukin-6. The exhausting time of cycling exercise challenge was not significantly increased in RES compared to that in placebo. However, IL-6 response was significantly decreased during exercise challenge in RES trial, and there were no differences in blood biomarkers, fatigue factors, and antioxidative response. Oral RES supplementation can attenuate exercise-induced IL-6 response but not fatigue and oxidative stress, inflammation response. However, we infer that 4-day oral RES supplementation has no ergogenic property on enhancing the high-intensity cycling exercise performance.


Assuntos
Ciclismo/fisiologia , Fadiga/diagnóstico , Interleucina-6/sangue , Substâncias para Melhoria do Desempenho/administração & dosagem , Resveratrol/administração & dosagem , Administração Oral , Adolescente , Atletas , Desempenho Atlético/fisiologia , Estudos Cross-Over , Fadiga/sangue , Fadiga/imunologia , Fadiga/prevenção & controle , Humanos , Inflamação/sangue , Inflamação/imunologia , Inflamação/prevenção & controle , Interleucina-6/metabolismo , Masculino , Estresse Oxidativo/efeitos dos fármacos , Método Simples-Cego , Resultado do Tratamento , Adulto Jovem
8.
Mol Neurodegener ; 15(1): 71, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33246484

RESUMO

BACKGROUND: Accumulation of amyloid-ß (Aß) peptide in the brain is a pathological hallmark of Alzheimer's disease (AD). The clusterin (CLU) gene confers a risk for AD and CLU is highly upregulated in AD patients, with the common non-coding, protective CLU variants associated with increased expression. Although there is strong evidence implicating CLU in amyloid metabolism, the exact mechanism underlying the CLU involvement in AD is not fully understood or whether physiologic alterations of CLU levels in the brain would be protective. RESULTS: We used a gene delivery approach to overexpress CLU in astrocytes, the major source of CLU expression in the brain. We found that CLU overexpression resulted in a significant reduction of total and fibrillar amyloid in both cortex and hippocampus in the APP/PS1 mouse model of AD amyloidosis. CLU overexpression also ameliorated amyloid-associated neurotoxicity and gliosis. To complement these overexpression studies, we also analyzed the effects of haploinsufficiency of Clu using heterozygous (Clu+/-) mice and control littermates in the APP/PS1 model. CLU reduction led to a substantial increase in the amyloid plaque load in both cortex and hippocampus in APP/PS1; Clu+/- mice compared to wild-type (APP/PS1; Clu+/+) littermate controls, with a concomitant increase in neuritic dystrophy and gliosis. CONCLUSIONS: Thus, both physiologic ~ 30% overexpression or ~ 50% reduction in CLU have substantial impacts on amyloid load and associated pathologies. Our results demonstrate that CLU plays a major role in Aß accumulation in the brain and suggest that efforts aimed at CLU upregulation via pharmacological or gene delivery approaches offer a promising therapeutic strategy to regulate amyloid pathology.


Assuntos
Amiloidose/metabolismo , Astrócitos/metabolismo , Clusterina/metabolismo , Placa Amiloide/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Camundongos Transgênicos , Placa Amiloide/patologia
9.
J Control Release ; 328: 87-99, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32858076

RESUMO

The combination of photothermal and photodynamic therapy (PTT/PDT) shows pronounced potential as a prominent therapeutic strategy for tumor treatment. However, the efficacy is limited by insufficient tumor-targeted delivery of PTT and PDT reagents and the hypoxic nature of the tumor microenvironment. To overcome these limitations, tumor acidity-responsive lipid membrane-enclosed perfluorooctyl bromide oil droplet nanoparticles (NPs) surface modified with N-acetyl histidine-modified D-α-tocopheryl polyethylene glycol 1000 succinate (PFOB@IMHNPs) were developed, capable of co-delivering oxygen, IR780 (a photothermal agent) and mTHPC (a photodynamic sensitizer) into tumors. Through self-sufficient oxygen transportation in combination with promotion of cellular uptake upon acid-triggered generation of surface positive charge, the PFOB@IMHNPs effectively delivered IR780 and mTHPC and produced singlet oxygen within hypoxic TRAMP-C1 cells following exposure to irradiation at 660 nm. This led to effective killing of hypoxic cancer cells in vitro. Importantly, when irradiation at 808 and 660 nm was carried out, PT/PD combination therapy utilizing PFOB@IMHNPs dramatically suppressed the growth of TRAMP-C1 tumors through effective tumor-targeted cargo delivery and relief of tumor hypoxia. Our results suggest the high potential of the PFOB@IMHNPs developed in this study in clinical application for cancer treatment.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Linhagem Celular Tumoral , Humanos , Neoplasias/tratamento farmacológico , Oxigênio , Fármacos Fotossensibilizantes/uso terapêutico , Microambiente Tumoral
10.
PLoS One ; 15(6): e0234549, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32555608

RESUMO

Methadone is a synthetic opioid used as maintenance treatment for patients addicted to heroin. Skin irritation is one of the adverse events caused by opioid use. 344 methadone maintenance treatment (MMT) patients were recruited with records and measurements on methadone dose, plasma methadone concentrations, and treatment emergent symptom scales (TESS). 15 patients reported with skin irritation. Five SNPs located within the NECTIN4 genetic region were genotyped. The NECTIN4 gene within the adherens junction interaction pathway was associated with methadone dose in pathway-based genome wide association analyses (P = 0.0008). Three highly-linked SNPs, rs11265549, rs3820097, and rs4656978, were significantly associated with methadone dose (P = 0.0003), plasma concentrations of R,S-methadone (P = 0.0004) and TNF-α (P = 0.010) in all 344 MMT patients, and with self-report skin irritation symptom scores (P = 0.010) in the 15 MMT patients who reported with skin irritation. To identify the possible roles of plasma level of Nectin-4 in the responses to MMT and opioid use, additional age- and gender-matched 51 controls and 83 methadone-free abstinent former heroin users were recruited. Plasma level of Nectin-4 was the highest in MMT patients among the three groups. The results suggest involvement of genetic variants on NECTIN4 in methadone dose. Plasma Nectin-4 level is likely an indicator for continued use of opioids.


Assuntos
Moléculas de Adesão Celular/genética , Dependência de Heroína/genética , Metadona/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/genética , Adulto , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Moléculas de Adesão Celular/sangue , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Dependência de Heroína/sangue , Dependência de Heroína/tratamento farmacológico , Dependência de Heroína/patologia , Humanos , Masculino , Metadona/efeitos adversos , Metadona/sangue , Tratamento de Substituição de Opiáceos/métodos , Transtornos Relacionados ao Uso de Opioides/sangue , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/patologia
11.
Sci Transl Med ; 12(529)2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32024798

RESUMO

The apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease mainly by driving amyloid-ß pathology. Recently, APOE4 has also been found to be a genetic risk factor for Lewy body dementia (LBD), which includes dementia with Lewy bodies and Parkinson's disease dementia. How APOE4 drives risk of LBD and whether it has a direct effect on α-synuclein pathology are not clear. Here, we generated a mouse model of synucleinopathy using an adeno-associated virus gene delivery of α-synuclein in human APOE-targeted replacement mice expressing APOE2, APOE3, or APOE4. We found that APOE4, but not APOE2 or APOE3, increased α-synuclein pathology, impaired behavioral performances, worsened neuronal and synaptic loss, and increased astrogliosis at 9 months of age. Transcriptomic profiling in APOE4-expressing α-synuclein mice highlighted altered lipid and energy metabolism and synapse-related pathways. We also observed an effect of APOE4 on α-synuclein pathology in human postmortem brains with LBD and minimal amyloid pathology. Our data demonstrate a pathogenic role of APOE4 in exacerbating α-synuclein pathology independent of amyloid, providing mechanistic insights into how APOE4 increases the risk of LBD.


Assuntos
Apolipoproteína E4 , Doença por Corpos de Lewy/genética , Sinucleinopatias , alfa-Sinucleína , Peptídeos beta-Amiloides , Animais , Apolipoproteína E4/genética , Camundongos , Camundongos Knockout para ApoE , Sinucleinopatias/genética
12.
Nat Commun ; 10(1): 1365, 2019 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-30911003

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial surface receptor genetically linked to the risk for Alzheimer's disease (AD). A proteolytic product, soluble TREM2 (sTREM2), is abundant in the cerebrospinal fluid and its levels positively correlate with neuronal injury markers. To gain insights into the pathological roles of sTREM2, we studied sTREM2 in the brain of 5xFAD mice, a model of AD, by direct stereotaxic injection of recombinant sTREM2 protein or by adeno-associated virus (AAV)-mediated expression. We found that sTREM2 reduces amyloid plaque load and rescues functional deficits of spatial memory and long-term potentiation. Importantly, sTREM2 enhances microglial proliferation, migration, clustering in the vicinity of amyloid plaques and the uptake and degradation of Aß. Depletion of microglia abolishes the neuroprotective effects of sTREM2. Our study demonstrates a protective role of sTREM2 against amyloid pathology and related toxicity and suggests that increasing sTREM2 can be explored for AD therapy.


Assuntos
Doença de Alzheimer/terapia , Potenciação de Longa Duração/efeitos dos fármacos , Glicoproteínas de Membrana/genética , Microglia/efeitos dos fármacos , Placa Amiloide/terapia , Receptores Imunológicos/genética , Memória Espacial/efeitos dos fármacos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Injeções Intraventriculares , Potenciação de Longa Duração/fisiologia , Masculino , Glicoproteínas de Membrana/administração & dosagem , Glicoproteínas de Membrana/metabolismo , Camundongos , Microglia/metabolismo , Microglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fenótipo , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Cultura Primária de Células , Proteólise , Receptores Imunológicos/administração & dosagem , Receptores Imunológicos/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Memória Espacial/fisiologia , Técnicas Estereotáxicas
13.
Nat Commun ; 9(1): 4388, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30348994

RESUMO

Apolipoprotein E (APOE) ε4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease mainly by modulating amyloid-ß pathology. APOE ε4 is also shown to exacerbate neurodegeneration and neuroinflammation in a tau transgenic mouse model. To further evaluate the association of APOE genotype with the presence and severity of tau pathology, we express human tau via an adeno-associated virus gene delivery approach in human APOE targeted replacement mice. We find increased hyperphosphorylated tau species, tau aggregates, and behavioral abnormalities in mice expressing APOE ε2/ε2. We also show that in humans, the APOE ε2 allele is associated with increased tau pathology in the brains of progressive supranuclear palsy (PSP) cases. Finally, we identify an association between the APOE ε2/ε2 genotype and risk of tauopathies using two series of pathologically-confirmed cases of PSP and corticobasal degeneration. Our data together suggest APOE ε2 status may influence the risk and progression of tauopathy.


Assuntos
Apolipoproteína E2/metabolismo , Tauopatias/metabolismo , Tauopatias/patologia , Alelos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Apolipoproteína E4/metabolismo , Progressão da Doença , Humanos , Camundongos , Paralisia Supranuclear Progressiva/genética , Paralisia Supranuclear Progressiva/metabolismo , Paralisia Supranuclear Progressiva/patologia
14.
Int J Neuropsychopharmacol ; 21(10): 910-917, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30060048

RESUMO

Background: There is no countable biomarker for opioid dependence treatment responses thus far. In this study, we recruited Taiwanese methadone maintenance treatment patients to search for genes involving the regulatory mechanisms of methadone dose by genome-wide association analyses. Methods: A total of 344 Taiwanese methadone maintenance treatment patients were included in a genome-wide association study. The involvement of GRK5 in opioid dependence was then further confirmed by gene expression study on lymphoblastoid cell lines derived from 3 independent age- and gender-matched groups: methadone maintenance treatment patients, medication-free former heroin abusers, and normal controls. Results: The results indicated that GRK5, the gene encoding an enzyme related to µ-opioid receptor desensitization, is associated with methadone dose by additive model of gene-based association analysis (P=6.76×10-5). We found that 6 of the 55 single nucleotide polymorphisms from the genome-wide genotype platform and 2 single nucleotide polymorphisms from the 29 additionally selected single nucleotide polymorphisms were significantly associated with methadone maintenance dose in both genotype and allele type (P ≤ .006), especially in patients who tested negative in the urine morphine test. The levels of GRK5 gene expression were similar between methadone maintenance treatment patients and medication-free former heroin abusers. However, the normal controls showed a significantly lower level of GRK5 gene expression than the other groups (P=.019). Conclusions: The results suggested an important role for GRK5 in the regulatory mechanisms of methadone dose and course of heroin dependence.


Assuntos
Quinase 5 de Receptor Acoplado a Proteína G/genética , Dependência de Heroína/genética , Metadona/uso terapêutico , Adulto , Estudos de Casos e Controles , Estudos Transversais , Feminino , Quinase 5 de Receptor Acoplado a Proteína G/biossíntese , Expressão Gênica , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Dependência de Heroína/tratamento farmacológico , Humanos , Masculino , Tratamento de Substituição de Opiáceos/métodos , Polimorfismo de Nucleotídeo Único/genética , Adulto Jovem
15.
Drug Alcohol Depend ; 183: 19-24, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29222992

RESUMO

BACKGROUND: Degeneration of central neurons and fibers has been observed in postmortem brains of heroin dependent patients. However, there are no biomarkers to predict the severity of neurodegeneration related to heroin dependence. A correlation has been reported between inflammatory C-C motif chemokine ligand 11 (CCL11, or eotaxin-1) and neurodegeneration in Alzheimer's disease. METHODS: Three-hundred-forty-four heroin dependent, Taiwanese patients under methadone maintenance treatment (MMT) were included with clinical assessment and genomics information. Eighty-seven normal control subjects were also recruited for comparison. RESULTS: Using receiver operating characteristics curve analyses, CCL11 showed the strongest sensitivity and specificity in correlation with age by a cut-off at 45 years (AUC = 0.69, P < 0.0001) in MMT patients, but not normal controls. Patients 45 years of age or older had significantly higher plasma levels of CCL11, fibroblast growth factor 2 (FGF-2), nicotine metabolite cotinine, and a longer duration of addiction. Plasma level of CCL11 was correlated with that of FGF-2 (partial r2 = 0.24, P < 0.0001). Carriers with the mutant allele of rs1129844, a functional single nucleotide polymorphism (Ala23Thr) in the CCL11 gene, showed a higher plasma level of Aß42, ratio of Aß42/Aß40, and insomnia side effect symptom score than the GG genotype carriers among MMT responders with morphine-negative urine results. CONCLUSIONS: The results suggest possible novel mechanisms mediated through CCL11 involving neurotoxicity in heroin dependent patients.


Assuntos
Envelhecimento/metabolismo , Quimiocina CCL11/genética , Dependência de Heroína/genética , Metadona/uso terapêutico , Adulto , Peptídeos beta-Amiloides/sangue , Encéfalo/fisiopatologia , Estudos de Casos e Controles , Quimiocina CCL11/sangue , Cotinina/sangue , Feminino , Fator 2 de Crescimento de Fibroblastos/sangue , Genótipo , Dependência de Heroína/sangue , Dependência de Heroína/complicações , Dependência de Heroína/tratamento farmacológico , Humanos , Masculino , Pessoa de Meia-Idade , Tratamento de Substituição de Opiáceos , Fragmentos de Peptídeos/sangue , Polimorfismo de Nucleotídeo Único/genética , Distúrbios do Início e da Manutenção do Sono/complicações , Distúrbios do Início e da Manutenção do Sono/genética
16.
Exp Neurol ; 300: 13-21, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29106980

RESUMO

Pericytes are a major component of cerebrovasculature playing a key role in maintaining cerebrovascular homeostasis. These cells have also been suggested to regulate brain metabolism of amyloid-ß (Aß), disturbances of which are believed to contribute to the pathogenesis of Alzheimer's disease (AD). To examine the effects of pericytes on brain Aß metabolism, C3H/10T1/2 mouse mesenchymal stem cells were differentiated into pericytes and stereotaxically injected into the brains of amyloid AD model APP/PS1 mice at the age of 18 to 20months. Consistent with a role of pericytes in modulating cerebrovascular function, brain microcirculation in the pericyte-injected hemisphere of the mice was increased 3weeks after implantation compared to the contralateral hemisphere when measured by laser speckle contrast analysis technology. Importantly, enzyme-linked immunosorbent assay revealed that the levels of insoluble Aß40 and Aß42 were significantly lower in the hippocampus of the pericyte-injected hemisphere of the APP/PS1 mice than that of the contralateral side. Consistently, immunohistochemical analysis demonstrated that the pericyte implantation reduced Aß deposition in the hippocampus. When brain slices from the APP/PS1 mice were incubated with C3H/10T1/2 cell-derived pericytes, Aß42 levels were significantly reduced in a manner that depends on the expression of a major Aß endocytic receptor, the low-density lipoprotein receptor-related protein 1 (LRP1). While LRP1 mediated the cellular uptake of Aß in the pericytes, the amounts of major Aß-degrading enzymes were not affected by LRP1 knockdown. Together, our findings indicate that mesenchymal stem cell-derived pericytes have the capacity to reduce brain Aß and related pathology, and suggest that cell-based therapy through transplantation of pericytes may be a promising approach to prevent and/or treat AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Circulação Cerebrovascular/fisiologia , Fragmentos de Peptídeos/metabolismo , Pericitos/fisiologia , Pericitos/transplante , Precursor de Proteína beta-Amiloide/genética , Animais , Encéfalo/irrigação sanguínea , Linhagem Celular Transformada , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Presenilina-1/genética
17.
Neuron ; 96(5): 1024-1032.e3, 2017 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-29216449

RESUMO

Accumulation and aggregation of amyloid-ß (Aß) in the brain is an initiating step in the pathogenesis of Alzheimer's disease (AD). The ε4 allele of apolipoprotein E (apoE) gene is the strongest genetic risk factor for late-onset AD. Although there is strong evidence showing that apoE4 enhances amyloid pathology, it is not clear what the critical stage(s) is during amyloid development in which apoE4 has the strongest impact. Using apoE inducible mouse models, we show that increased expression of astrocytic apoE4, but not apoE3, during the seeding stage of amyloid development enhanced amyloid deposition and neuritic dystrophy in amyloid model mice. ApoE4, but not apoE3, significantly increased brain Aß half-life measured by in vivo microdialysis. Furthermore, apoE4 expression increased whereas apoE3 reduced amyloid-related gliosis in the mouse brains. Together, our results demonstrate that apoE4 has the greatest impact on amyloid during the seeding stage, likely by perturbing Aß clearance and enhancing Aß aggregation.


Assuntos
Amiloidose/patologia , Apolipoproteína E4/farmacologia , Doença de Alzheimer/patologia , Amiloidose/genética , Animais , Apolipoproteína E3/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/patologia , Técnicas de Introdução de Genes , Gliose/patologia , Humanos , Camundongos , Camundongos Transgênicos , Neuritos/efeitos dos fármacos , Neuritos/patologia
18.
Proc Natl Acad Sci U S A ; 114(33): E6962-E6971, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28701379

RESUMO

Alzheimer's disease (AD) is characterized by amyloid-ß (Aß) peptide deposition in brain parenchyma as plaques and in cerebral blood vessels as cerebral amyloid angiopathy (CAA). CAA deposition leads to several clinical complications, including intracerebral hemorrhage. The underlying molecular mechanisms that regulate plaque and CAA deposition in the vast majority of sporadic AD patients remain unclear. The clusterin (CLU) gene is genetically associated with AD and CLU has been shown to alter aggregation, toxicity, and blood-brain barrier transport of Aß, suggesting it might play a key role in regulating the balance between Aß deposition and clearance in both brain and blood vessels. Here, we investigated the effect of CLU on Aß pathology using the amyloid precursor protein/presenilin 1 (APP/PS1) mouse model of AD amyloidosis on a Clu+/+ or Clu-/- background. We found a marked decrease in plaque deposition in the brain parenchyma but an equally striking increase in CAA within the cerebrovasculature of APP/PS1;Clu-/- mice. Surprisingly, despite the several-fold increase in CAA levels, APP/PS1;Clu-/- mice had significantly less hemorrhage and inflammation. Mice lacking CLU had impaired clearance of Aß in vivo and exogenously added CLU significantly prevented Aß binding to isolated vessels ex vivo. These findings suggest that in the absence of CLU, Aß clearance shifts to perivascular drainage pathways, resulting in fewer parenchymal plaques but more CAA because of loss of CLU chaperone activity, complicating the potential therapeutic targeting of CLU for AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Clusterina/deficiência , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/patologia , Modelos Animais de Doenças , Camundongos , Camundongos Mutantes
19.
J Neurosci ; 37(15): 4023-4031, 2017 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-28275161

RESUMO

Accumulation and deposition of amyloid-ß (Aß) in the brain represent an early and perhaps necessary step in the pathogenesis of Alzheimer's disease (AD). Aß accumulation leads to the formation of Aß aggregates, which may directly and indirectly lead to eventual neurodegeneration. While Aß production is accelerated in many familial forms of early-onset AD, increasing evidence indicates that impaired clearance of Aß is more evident in late-onset AD. To uncover the mechanisms underlying impaired Aß clearance in AD, we examined the role of low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. Although LRP1 has been shown to play critical roles in brain Aß metabolism in neurons and vascular mural cells, its role in astrocytes, the most abundant cell type in the brain responsible for maintaining neuronal homeostasis, remains unclear. Here, we show that astrocytic LRP1 plays a critical role in brain Aß clearance. LRP1 knockdown in primary astrocytes resulted in decreased cellular Aß uptake and degradation. In addition, silencing of LRP1 in astrocytes led to downregulation of several major Aß-degrading enzymes, including matrix metalloproteases MMP2, MMP9, and insulin-degrading enzyme. More important, conditional knock-out of the Lrp1 gene in astrocytes in the background of APP/PS1 mice impaired brain Aß clearance, exacerbated Aß accumulation, and accelerated amyloid plaque deposition without affecting its production. Together, our results demonstrate that astrocytic LRP1 plays an important role in Aß metabolism and that restoring LRP1 expression and function in the brain could be an effective strategy to facilitate Aß clearance and counter amyloid pathology in AD.SIGNIFICANCE STATEMENT Astrocytes represent a major cell type regulating brain homeostasis; however, their roles in brain clearance of amyloid-ß (Aß) and underlying mechanism are not clear. In this study, we used both cellular models and conditional knock-out mouse models to address the role of a critical Aß receptor, the low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. We found that LRP1 in astrocytes plays a critical role in brain Aß clearance by modulating several Aß-degrading enzymes and cellular degradation pathways. Our results establish a critical role of astrocytic LRP1 in brain Aß clearance and shed light on specific Aß clearance pathways that may help to establish new targets for AD prevention and therapy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/metabolismo , Receptores de LDL/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Astrócitos/patologia , Encéfalo/patologia , Células Cultivadas , Feminino , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Masculino , Camundongos , Camundongos Knockout
20.
J Exp Med ; 214(3): 597-607, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28209725

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor expressed in microglia in the brain. A soluble form of TREM2 (sTREM2) derived from proteolytic cleavage of the cell surface receptor is increased in the preclinical stages of AD and positively correlates with the amounts of total and phosphorylated tau in the cerebrospinal fluid. However, the physiological and pathological functions of sTREM2 remain unknown. Here, we show that sTREM2 promotes microglial survival in a PI3K/Akt-dependent manner and stimulates the production of inflammatory cytokines depending on NF-κB. Variants of sTREM2 carrying AD risk-associated mutations were less potent in both suppressing apoptosis and triggering inflammatory responses. Importantly, sTREM2 delivered to the hippocampi of both wild-type and Trem2-knockout mice elevated the expression of inflammatory cytokines and induced morphological changes of microglia. Collectively, these data indicate that sTREM2 triggers microglial activation inducing inflammatory responses and promoting survival. This study has implications for the pathogenesis of AD and provides insights into targeting sTREM2 pathway for AD therapy.


Assuntos
Inflamação/etiologia , Glicoproteínas de Membrana/fisiologia , Microglia/fisiologia , Receptores Imunológicos/fisiologia , Doença de Alzheimer/etiologia , Animais , Sobrevivência Celular , Glicogênio Sintase Quinase 3 beta/fisiologia , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , NF-kappa B/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Imunológicos/genética , Transdução de Sinais , beta Catenina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA