Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neuroinflammation ; 18(1): 272, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34798897

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by pathological deposition of misfolded self-protein amyloid beta (Aß) which in kind facilitates tau aggregation and neurodegeneration. Neuroinflammation is accepted as a key disease driver caused by innate microglia activation. Recently, adaptive immune alterations have been uncovered that begin early and persist throughout the disease. How these occur and whether they can be harnessed to halt disease progress is unclear. We propose that self-antigens would induct autoreactive effector T cells (Teffs) that drive pro-inflammatory and neurodestructive immunity leading to cognitive impairments. Here, we investigated the role of effector immunity and how it could affect cellular-level disease pathobiology in an AD animal model. METHODS: In this report, we developed and characterized cloned lines of amyloid beta (Aß) reactive type 1 T helper (Th1) and type 17 Th (Th17) cells to study their role in AD pathogenesis. The cellular phenotype and antigen-specificity of Aß-specific Th1 and Th17 clones were confirmed using flow cytometry, immunoblot staining and Aß T cell epitope loaded haplotype-matched major histocompatibility complex II IAb (MHCII-IAb-KLVFFAEDVGSNKGA) tetramer binding. Aß-Th1 and Aß-Th17 clones were adoptively transferred into APP/PS1 double-transgenic mice expressing chimeric mouse/human amyloid precursor protein and mutant human presenilin 1, and the mice were assessed for memory impairments. Finally, blood, spleen, lymph nodes and brain were harvested for immunological, biochemical, and histological analyses. RESULTS: The propagated Aß-Th1 and Aß-Th17 clones were confirmed stable and long-lived. Treatment of APP/PS1 mice with Aß reactive Teffs accelerated memory impairment and systemic inflammation, increased amyloid burden, elevated microglia activation, and exacerbated neuroinflammation. Both Th1 and Th17 Aß-reactive Teffs progressed AD pathology by downregulating anti-inflammatory and immunosuppressive regulatory T cells (Tregs) as recorded in the periphery and within the central nervous system. CONCLUSIONS: These results underscore an important pathological role for CD4+ Teffs in AD progression. We posit that aberrant disease-associated effector T cell immune responses can be controlled. One solution is by Aß reactive Tregs.


Assuntos
Doença de Alzheimer/patologia , Linfócitos T CD4-Positivos/patologia , Presenilina-1/genética , Precursor de Proteína beta-Amiloide/genética , Amiloidose/patologia , Animais , Transtornos Cognitivos/patologia , Transtornos Cognitivos/psicologia , Inflamação/genética , Camundongos , Camundongos Transgênicos , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th1/patologia , Células Th17/imunologia , Células Th17/patologia
2.
J Neuroinflammation ; 15(1): 137, 2018 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-29729668

RESUMO

BACKGROUND: The mixed lineage kinase type 3 inhibitor URMC-099 facilitates amyloid-beta (Aß) clearance and degradation in cultured murine microglia. One putative mechanism is an effect of URMC-099 on Aß uptake and degradation. As URMC-099 promotes endolysosomal protein trafficking and reduces Aß microglial pro-inflammatory activities, we assessed whether these responses affect Aß pathobiogenesis. To this end, URMC-099's therapeutic potential, in Aß precursor protein/presenilin-1 (APP/PS1) double-transgenic mice, was investigated in this model of Alzheimer's disease (AD). METHODS: Four-month-old APP/PS1 mice were administered intraperitoneal URMC-099 injections at 10 mg/kg daily for 3 weeks. Brain tissues were examined by biochemical, molecular and immunohistochemical tests. RESULTS: URMC-099 inhibited mitogen-activated protein kinase 3/4-mediated activation and attenuated ß-amyloidosis. Microglial nitric oxide synthase-2 and arginase-1 were co-localized with lysosomal-associated membrane protein 1 (Lamp1) and Aß. Importatly, URMC-099 restored synaptic integrity and hippocampal neurogenesis in APP/PS1 mice. CONCLUSIONS: URMC-099 facilitates Aß clearance in the brain of APP/PS1 mice. The multifaceted immune modulatory and neuroprotective roles of URMC-099 make it an attractive candidate for ameliorating the course of AD. This is buttressed by removal of pathologic Aß species and restoration of the brain's microenvironment during disease.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Modelos Animais de Doenças , Piridinas/uso terapêutico , Pirróis/uso terapêutico , Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide/genética , Animais , Células Cultivadas , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Presenilina-1/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/farmacologia , Pirróis/farmacologia
3.
J Neuroimmunol ; 319: 80-92, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29573847

RESUMO

We investigated the effects of granulocyte-macrophage colony stimulating factor (GM-CSF) on behavioral and pathological outcomes in Alzheimer's disease (AD) and non-transgenic mice. GM-CSF treatment in AD mice reduced brain amyloidosis, increased plasma Aß, and rescued cognitive impairment with increased hippocampal expression of calbindin and synaptophysin and increased levels of doublecortin-positive cells in the dentate gyrus. These data extend GM-CSF pleiotropic neuroprotection mechanisms in AD and include regulatory T cell-mediated immunomodulation of microglial function, Aß clearance, maintenance of synaptic integrity, and induction of neurogenesis. Together these data support further development of GM-CSF as a neuroprotective agent for AD.


Assuntos
Doença de Alzheimer , Encéfalo/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Camundongos , Camundongos Transgênicos
4.
J Neuroimmune Pharmacol ; 12(2): 340-352, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27966067

RESUMO

Amyloid-ß (Aß) precursor protein (APP) metabolism engages neuronal endolysosomal pathways for Aß processing and secretion. In Alzheimer's disease (AD), dysregulation of APP leads to excess Aß and neuronal dysfunction; suggesting that neuronal APP/Aß trafficking can be targeted for therapeutic gain. Cathepsin B (CatB) is a lysosomal cysteine protease that can lower Aß levels. However, whether CatB-modulation of Aß improves learning and memory function deficits in AD is not known. To this end, progenitor neurons were infected with recombinant adenovirus expressing CatB and recovered cell lysates subjected to proteomic analyses. The results demonstrated Lamp1 deregulation and linkages between CatB and the neuronal phagosome network. Hippocampal injections of adeno-associated virus expressing CatB reduced Aß levels, increased Lamp1 and improved learning and memory. The findings were associated with the emergence of c-fos + cells. The results support the idea that CatB can speed Aß metabolism through lysosomal pathways and as such reduce AD-associated memory deficits.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Precursor de Proteína beta-Amiloide , Amiloidose/tratamento farmacológico , Catepsina B/uso terapêutico , Aprendizagem/efeitos dos fármacos , Memória/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/patologia , Animais , Catepsina B/farmacologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Aprendizagem/fisiologia , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Presenilina-1/metabolismo
5.
J Neuroinflammation ; 13(1): 184, 2016 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-27401058

RESUMO

BACKGROUND: Amyloid-ß (Aß)-stimulated microglial inflammatory responses engage mitogen-activated protein kinase (MAPK) pathways in Alzheimer's disease (AD). Mixed-lineage kinases (MLKs) regulate upstream MAPK signaling that include p38 MAPK and c-Jun amino-terminal kinase (JNK). However, whether MLK-MAPK pathways affect Aß-mediated neuroinflammation is unknown. To this end, we investigated if URMC-099, a brain-penetrant small-molecule MLK type 3 inhibitor, can modulate Aß trafficking and processing required for generating AD-associated microglial inflammatory responses. METHODS: Aß1-42 (Aß42) and/or URMC-099-treated murine microglia were investigated for phosphorylated mitogen-activated protein kinase kinase (MKK)3, MKK4 (p-MKK3, p-MKK4), p38 (p-p38), and JNK (p-JNK). These pathways were studied in tandem with the expression of the pro-inflammatory cytokines interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α. Gene expression of the anti-inflammatory cytokines, IL-4 and IL-13, was evaluated by real-time quantitative polymerase chain reaction. Aß uptake and expression of scavenger receptors were measured. Protein trafficking was assessed by measures of endolysosomal markers using confocal microscopy. RESULTS: Aß42-mediated microglial activation pathways were shown by phosphorylation of MKK3, MKK4, p38, and JNK and by expression of IL-1ß, IL-6, and TNF-α. URMC-099 modulated microglial inflammatory responses with induction of IL-4 and IL-13. Phagocytosis of Aß42 was facilitated by URMC-099 with up-regulation of scavenger receptors. Co-localization of Aß and endolysosomal markers associated with enhanced Aß42 degradation was observed. CONCLUSIONS: URMC-099 reduced microglial inflammatory responses and facilitated phagolysosomal trafficking with associated Aß degradation. These data demonstrate a new immunomodulatory role for URMC-099 to inhibit MLK and to induce microglial anti-inflammatory responses. Thus, URMC-099 may be developed further as a novel disease-modifying AD therapy.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Citocinas/metabolismo , Microglia/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Pirróis/farmacologia , Peptídeos beta-Amiloides/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Células Cultivadas , Citocinas/genética , Ensaio de Imunoadsorção Enzimática , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Microscopia Confocal , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fragmentos de Peptídeos/farmacologia , Receptores Depuradores/genética , Receptores Depuradores/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estatísticas não Paramétricas , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
6.
Neurobiol Aging ; 36(11): 2995-3007, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26315370

RESUMO

Cluster of Differentiation-200 (CD200) is an anti-inflammatory glycoprotein expressed in neurons, T cells, and B cells, and its receptor is expressed on glia. Both Alzheimer's disease patients and mouse models display age-related or amyloid-ß peptide (Aß)-induced reductions in CD200. The goal of this study was to determine if neuronal CD200 expression restores hippocampal neurogenesis and reduces Aß in the amyloid precursor protein mouse model. Amyloid precursor protein and wild-type mice were injected at 6 months of age with an adeno-associated virus expressing CD200 into the hippocampus and sacrificed at 12 months. CD200 expression restored neural progenitor cell proliferation and differentiation in the subgranular and granular cell layers of the dentate gyrus and reduced diffuse but not thioflavin-S(+) plaques in the hippocampus. In vitro studies demonstrated that CD200-stimulated microglia increased neural differentiation of neural stem cells and enhanced axon elongation and dendrite number. CD200 also enhanced Aß uptake by microglia. These data indicate that CD200 is capable of enhancing microglia-mediated Aß clearance and neural differentiation and has potential as a therapeutic for Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Antígenos CD/fisiologia , Hipocampo/fisiologia , Neurogênese/genética , Fagocitose/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Antígenos CD/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Hipocampo/citologia , Camundongos Transgênicos , Microglia/citologia , Microglia/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/efeitos dos fármacos , Células-Tronco/citologia
7.
Mol Ther ; 23(11): 1712-1721, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26227349

RESUMO

Modulation of the amyloid-ß (Aß) trafficking pathway heralds a new therapeutic frontier for Alzheimer's disease (AD). As CD74 binds to the amyloid-ß precursor protein (APP) and can suppresses Aß processing, we investigated whether recombinant adeno-associated virus (AAV) delivery of CD74 could reduce Aß production and affect disease outcomes. This idea was tested in a mouse AD model. Cotransduction of AAV-tetracycline-controlled transactivator (tTA) and AAV-tet-response element (TRE)-CD74 resulted in CD74 expression, reduced Aß production in mouse neurons containing the human APP with familial AD-linked mutations. Stereotaxic injection of AAV-TRE-GFP or CD74 into the hippocampi of an AD mouse, defined as a TgCRND8 × calmodulin-dependent protein kinase II derived promoter-tTA double-transgenic, reduced Aß loads and pyramidal neuronal Aß accumulation in the hippocampus. Immunofluorescent studies showed that APP colocalization with Lamp1 was increased in CD74-expressing neurons. Moreover, Morris water maze tasks demonstrated that mice treated with AAV-TRE-CD74 showed improved learning and memory compared to AAV-TRE-GFP control animals. These results support the idea that CD74-induced alteration of Aß processing could improve AD-associated memory deficits as shown in mouse models of human disease.


Assuntos
Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/metabolismo , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Histocompatibilidade Classe II/genética , Neurônios/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Precursor de Proteína beta-Amiloide/fisiologia , Amiloidose/metabolismo , Animais , Células Cultivadas , Dependovirus , Modelos Animais de Doenças , Feminino , Terapia Genética , Vetores Genéticos/uso terapêutico , Hipocampo/metabolismo , Hipocampo/cirurgia , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Masculino , Aprendizagem em Labirinto , Memória , Camundongos , Camundongos Transgênicos , Microinjeções/métodos , Técnicas Estereotáxicas
8.
Brain Behav Immun ; 49: 311-21, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26112421

RESUMO

Aberrations in hippocampal neurogenesis are associated with learning and memory, synaptic plasticity and neurodegeneration in Alzheimer's disease (AD). However, the linkage between them, ß-amyloidosis and neuroinflammation is not well understood. To this end, we generated a mouse overexpressing familial AD (FAD) mutant human presenilin-1 (PS1) crossed with a knockout (KO) of the CC-chemokine ligand 2 (CCL2) gene. The PS1/CCL2KO mice developed robust age-dependent deficits in hippocampal neurogenesis associated with impairments in learning and memory, synaptic plasticity and long-term potentiation. Neurogliogenesis gene profiling supported ß-amyloid independent pathways for FAD-associated deficits in hippocampal neurogenesis. We conclude that these PS1/CCL2KO mice are suitable for studies linking host genetics, immunity and hippocampal function.


Assuntos
Doença de Alzheimer/fisiopatologia , Quimiocina CCL2/genética , Modelos Animais de Doenças , Hipocampo/fisiopatologia , Neurogênese , Presenilina-1/genética , Doença de Alzheimer/genética , Doença de Alzheimer/psicologia , Animais , Quimiocina CCL2/metabolismo , Hipocampo/metabolismo , Humanos , Potenciação de Longa Duração/genética , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/metabolismo , Neurônios/fisiologia , Presenilina-1/metabolismo
9.
J Alzheimers Dis ; 44(1): 297-307, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25227315

RESUMO

BACKGROUND: Redox-active metal dyshomeostasis and oxidative stress are associated with mitochondrial dysfunction and amyloid-ß (Aß) neurotoxicity that are linked to both the development of age-related macular degeneration (AMD) and Alzheimer's disease (AD). As potential therapeutic agents, orally active multifunctional antioxidants (MFAOs) possessing two independent functional groups capable of binding redox-active metals and scavenging free radicals have been synthesized. OBJECTIVE: To determine whether MFAOs affect mitochondrial function and reduce the presence of Aß plaque formation. METHODS: The MFAOs were evaluated in cultured SH-SY5Y cells and ARPE-19 cells. MFAO effects on mitochondrial function were investigated using rhodamine 123 staining after 2 hour exposure to MnCl2. MFAO effects on Aß:Zn complex formation were evaluated with Zinquin staining and the ability of the Aß:Zn complex to be degraded by matrix metalloproteinase-2 (MMP-2). The ability of MFAOs to reduce Aß plaque in the brain was determined by orally feeding MFAO for one year to B6;129-Psen1tm1Mpm Tg(AßPPSwe,tauP301L) 1Lfa/Mmjax transgenic mice. Aß levels were determined by ELISA. RESULTS: MFAOs neither adversely affected mitochondrial signaling nor labile cytoplasmic zinc levels. MFAOs protected cells against Mn2+-induced mitochondrial dysfunction. MFAOs also removed zinc from the Aß:Zn complex so that Aß plaque could be degraded by MMP-2. Zinquin staining indicated that the removed zinc was present in the cytoplasm as labile zinc. Orally administered MFAOs reduced the brain levels of both Aß40 and Aß42 isoforms of Aß. CONCLUSION: These studies demonstrate that these MFAOs have metal attenuating properties with therapeutic potential in the treatment of both AMD and AD.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Mitocôndrias/efeitos dos fármacos , Zinco/metabolismo , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Corantes Fluorescentes , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neuroblastoma/patologia , Quinolonas , Rodamina 123 , Compostos de Tosil
10.
Neurobiol Aging ; 34(4): 1060-8, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23040664

RESUMO

Neuroinflammation affects the pathobiology of Alzheimer's disease (AD). Notably, ß-amyloid (Aß) deposition induces microglial activation and the subsequent production of proinflammatory neurotoxic factors. In maintaining brain homeostasis, microglial plasticity also enables phenotypic transition between toxic and trophic activation states. One important control for such cell activation is through the CC-chemokine ligand 2 (CCL2) and its receptor, the CC-chemokine receptor 2. Both affect microglia and peripheral macrophage immune responses and for the latter, cell ingress across the blood-brain barrier. However, how CCL2-CC-chemokine receptor 2 signaling contributes to AD pathogenesis is not well understood. To this end, we now report that CCL2 deficiency influences behavioral abnormalities and disease progression in Aß precursor protein/presenilin-1 double-transgenic mice. Here, increased cortical and hippocampal Aß deposition is coincident with the formulation of Aß oligomers. Deficits in peripheral Aß clearance and in scavenger, neuroprogenitor, and microglial cell functions are linked to deficient Aß uptake. All serve to accelerate memory dysfunction. Taken together, these data support a role of CCL2 in innate immune functions relevant to AD pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Quimiocina CCL2/metabolismo , Transtornos Cognitivos/metabolismo , Modelos Animais de Doenças , Doença de Alzheimer/complicações , Amiloidose/complicações , Amiloidose/metabolismo , Animais , Transtornos Cognitivos/complicações , Camundongos , Camundongos Transgênicos
11.
Neurosci Lett ; 523(2): 167-73, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22776646

RESUMO

Inducible nitric oxide synthase (iNOS) upregulation and consequent NO formation are well-recognized neuroinflammatory responses associated with Parkinson's disease (PD). These contribute to nitrosative protein modifications affecting neuronal injury and cell death. Indeed, a pathobiologic signature for PD is Lewy body formation containing misfolded and aggregated forms of alpha-synuclein (α-syn). Moreover, nitration of α-syn promotes protein aggregation in disease. To model such pathological events, we constructed controllable iNOS and bicistronic α-syn-IRES-tTA adeno-associated virus (AAV) expression vectors. Transduction of iNOS and α-syn AAV constructs led to nitration of α-syn in neurons and overexpression of iNOS promoted protein aggregation. We posit that this AAV system mimics critical protein misfolding events associated with the pathogenesis of PD.


Assuntos
Óxido Nítrico/metabolismo , Doença de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Dependovirus/genética , Células HEK293 , Humanos , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Dobramento de Proteína , Transdução Genética , alfa-Sinucleína/genética
12.
J Neuroimmune Pharmacol ; 7(2): 412-23, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21826404

RESUMO

Combined antiretroviral therapy (ART) tremendously improved the lifespan and symptoms associated with AIDS-defining illness in affected individuals. However, chronic ART-treated patients frequently develop age-dependent complications, including dementia, diabetes, and hyperlipidemia: all risk factors of Alzheimer's disease. Importantly, the effect of ART compounds on amyloid generation and clearance has never been systematically examined. Nine prescribed HIV protease inhibitors were tested for their effect on amyloid-ß peptide (Aß) clearance in primary cultured human monocyte-derived macrophages. Atazanavir, ritonavir, and saquinavir modestly inhibited of Aß degradation, while lopinavir, nelfinavir, and ritonavir enhanced secretion of undigested Aß after phagocytosis. Lopinavir, nelfinavir, ritonavir, and saquinavir inhibited endogenous Aß40 production from primary cultured human cortical neurons, which were associated with reduction in Beta-site APP Converting Enzyme 1 (BACE1) and γ-secretase enzyme activities. However, ART compounds showed little inhibition of purified BACE1 activity in vitro, suggesting the indirect effect of ART compounds on BACE1 activity in neurons. Finally, nefinavir or lopinavir/ritonavir (Kaletra) were orally administered for 30 days into APP SCID mice expressing a double mutant form of APP 695 (KM670/671NL + V717F) in homozygosity for the scid allele of Prkdc. There was no difference in beta-amyloidosis by ART drug administration as determined by both immunohistochemistry and ELISA measurements although the therapeutic doses of the ART compounds was present in the brain. These data demonstrated that ART drugs can inhibit Aß clearance in macrophages and Aß production in neurons, but these effects did not significantly alter Aß accumulation in the mouse brain.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/efeitos dos fármacos , Peptídeos beta-Amiloides/metabolismo , Inibidores da Protease de HIV/farmacologia , Doença de Alzheimer/patologia , Animais , Sulfato de Atazanavir , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Imuno-Histoquímica , Lopinavir/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos SCID , Camundongos Transgênicos , Nelfinavir/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oligopeptídeos/farmacologia , Piridinas/farmacologia , Ritonavir/farmacologia , Saquinavir/farmacologia
13.
Proc Natl Acad Sci U S A ; 108(49): E1339-48, 2011 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-22042871

RESUMO

The adult hippocampus plays a central role in memory formation, synaptic plasticity, and neurogenesis. The subgranular zone of the dentate gyrus contains neural progenitor cells with self-renewal and multilineage potency. Transgene expression of familial Alzheimer's disease-linked mutants of ß-amyloid precursor protein (APP) and presenilin-1 leads to a significant inhibition of neurogenesis, which is potentially linked to age-dependent memory loss. To investigate the effect of neurogenesis on cognitive function in a relevant disease model, FGF2 gene is delivered bilaterally to the hippocampi of APP+presenilin-1 bigenic mice via an adenoassociated virus serotype 2/1 hybrid (AAV2/1-FGF2). Animals injected with AAV2/1-FGF2 at a pre- or postsymptomatic stage show significantly improved spatial learning in the radial arm water maze test. A neuropathological investigation demonstrates that AAV2/1-FGF2 injection enhances the number of doublecortin, BrdU/NeuN, and c-fos-positive cells in the dentate gyrus, and the clearance of fibrillar amyloid-ß peptide (Aß) in the hippocampus. AAV2/1-FGF2 injection also enhances long-term potentiation in another APP mouse model (J20) compared with control AAV2/1-GFP-injected littermates. An in vitro study confirmed the enhanced neurogenesis of mouse neural stem cells by direct AAV2/1-FGF2 infection in an Aß oligomer-sensitive manner. Further, FGF2 enhances Aß phagocytosis in primary cultured microglia, and reduces Aß production from primary cultured neurons after AAV2/1-FGF2 infection. Thus, our data indicate that virus-mediated FGF2 gene delivery has potential as an alternative therapy of Alzheimer's disease and possibly other neurocognitive disorders.


Assuntos
Doença de Alzheimer/metabolismo , Transtornos Cognitivos/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hipocampo/metabolismo , Doenças Neurodegenerativas/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , Transtornos Cognitivos/genética , Transtornos Cognitivos/terapia , Giro Denteado/metabolismo , Giro Denteado/patologia , Dependovirus/genética , Modelos Animais de Doenças , Fator 2 de Crescimento de Fibroblastos/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Hipocampo/patologia , Hipocampo/fisiopatologia , Humanos , Imuno-Histoquímica , Potenciação de Longa Duração/fisiologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/terapia , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo
14.
J Immunol ; 186(12): 6925-32, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21551363

RESUMO

The advent and wide introduction of antiretroviral therapy has greatly improved the survival and longevity of HIV-infected patients. Unfortunately, despite antiretroviral therapy treatment, these patients are still afflicted with many complications including cognitive dysfunction. There is a growing body of reports indicating accelerated deposition of amyloid plaques, which are composed of amyloid-ß peptide (Aß), in HIV-infected brains, though how HIV viral infection precipitates Aß accumulation is poorly understood. It is suggested that viral infection leads to increased production and impaired degradation of Aß. Mononuclear phagocytes (macrophages and microglia) that are productively infected by HIV in brains play a pivotal role in Aß degradation through the expression and execution of two endopeptidases, neprilysin (NEP) and insulin-degrading enzyme. In this study, we report that NEP has the dominant endopeptidase activity toward Aß in macrophages. Further, we demonstrate that monomeric Aß degradation by primary cultured macrophages and microglia was significantly impaired by HIV infection. This was accompanied with great reduction of NEP endopeptidase activity, which might be due to the diminished transport of NEP to the cell surface and intracellular accumulation at the endoplasmic reticulum and lysosomes. Therefore, these data suggest that malfunction of NEP in infected macrophages may contribute to acceleration of ß amyloidosis in HIV-inflicted brains, and modulation of macrophages may be a potential preventative target of Aß-related cognitive disorders in HIV-affected patients.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Transtornos Cognitivos/etiologia , Infecções por HIV/complicações , Neprilisina/metabolismo , Fagócitos/virologia , Células Cultivadas , Transtornos Cognitivos/virologia , Endopeptidases/metabolismo , HIV-1/fisiologia , Humanos , Insulisina/metabolismo , Fagócitos/enzimologia
15.
FASEB J ; 24(8): 3093-102, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20371618

RESUMO

Cytokines play an emerging role as neurotransmitters, neuromodulators, and neurohormones in the brain. This paradigm shift in cytokine function offers a new framework to understand their roles in ameliorating neurodegenerative disorders, such as Alzheimer's disease (AD). Molecular adjuvant therapy of AD animal models with glatiramer acetate induces anti-inflammatory responses and therapeutic effects. Although these effects are potentially mediated through anti-inflammatory cytokine signaling, the exact molecular identities and pathways are poorly understood. Here, we show that virus-mediated expression of the mouse interleukin (IL)-4 gene in beta-amyloid precursor protein + presenilin-1 (APP+PS1) bigenic mice attenuates AD pathogenesis. Introduction of an adeno-associated viral (AAV) vector encoding IL-4 into the hippocampus resulted in sustained expression of IL-4, reduced astro/microgliosis, amyloid-beta peptide (Abeta) oligomerization and deposition, and enhanced neurogenesis. Moreover, increased levels of IL-4 improved spatial learning, promoted phosphorylation of N-methyl-D-aspartate receptor subunit 2B at Tyr 1472, and enhanced its cell surface retention both in vivo and in vitro. Our data suggest that neuronal anti-inflammatory cytokine signaling may be a potential alternative target for non-Abeta-mediated treatment of AD.


Assuntos
Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Sistema Nervoso Central/metabolismo , Interleucina-4/biossíntese , Presenilina-1/genética , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Terapia Genética , Hipocampo/metabolismo , Interleucina-4/genética , Interleucina-4/farmacologia , Camundongos , Camundongos Mutantes , Neurônios/química , Neurônios/metabolismo , Transdução de Sinais , Transfecção
16.
PLoS One ; 4(7): e6197, 2009 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-19593388

RESUMO

BACKGROUND: The linkages between neuroinflammation and Alzheimer's disease (AD) pathogenesis are well established. What is not, however, is how specific immune pathways and proteins affect the disease. To this end, we previously demonstrated that transgenic over-expression of CCL2 enhanced microgliosis and induced diffuse amyloid plaque deposition in Tg2576 mice. This rodent model of AD expresses a Swedish beta-amyloid (Abeta) precursor protein mutant. METHODOLOGY/PRINCIPAL FINDINGS: We now report that CCL2 transgene expression accelerates deficits in spatial and working memory and hippocampal synaptic transmission in beta-amyloid precursor protein (APP) mice as early as 2-3 months of age. This is followed by increased numbers of microglia that are seen surrounding Abeta oligomers. CCL2 does not suppress Abeta degradation. Rather, CCL2 and tumor necrosis factor-alpha directly facilitated Abeta uptake, intracellular Abeta oligomerization, and protein secretion. CONCLUSIONS/SIGNIFICANCE: We posit that CCL2 facilitates Abeta oligomer formation in microglia and propose that such events accelerate memory dysfunction by affecting Abeta seeding in the brain.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Biopolímeros/biossíntese , Quimiocina CCL2/fisiologia , Transtornos Cognitivos/fisiopatologia , Microglia/fisiologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Biopolímeros/metabolismo , Western Blotting , Quimiocina CCL2/genética , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Transgênicos , Fator de Necrose Tumoral alfa/fisiologia
17.
Biochim Biophys Acta ; 1790(5): 326-38, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19364479

RESUMO

BACKGROUND: Fortilin negatively regulates apoptosis and is overexpressed in cancer. However, the role of fortilin in mammalian development is not clear. METHODS AND RESULTS: In order to evaluate the physiological role of fortilin in vivo, we performed a targeted disruption of the fortilin gene in mice. Fortilin(+/-) mice have the ability to survive and exhibit normal growth, while fortilin(-/-) mice are embryonically lethal around the 3.5 days post-coital (dpc). Cultured blastocysts from fortilin(+/-) embryos undergo normal outgrowth to produce inner cell mass (ICM) and trophoblasts (TB), while ICM of fortilin(-/-) embryos either fails to outgrow or prematurely disintegrates. Mouse embryonic fibroblasts (MEF) derived from fortilin(+/-) embryos are more susceptible to noxious stimuli than are wild type embryos. It has been consistently shown in Xenopus embryos that the depletion of fortilin's message severely compromises the formation of neural tissue, even in the brain, while overexpression of fortilin induces the partial double body axis in embryos and is capable of blocking BMP4-induced transcription of Vent1, Vent2, and Msx1 genes. This suggests that fortilin is an inhibitor of the BMP pathway. Strikingly, when fortilin levels are reduced by siRNA, BMP4 causes MEF to undergo extensive DNA-fragmentation, while DNA fragmentation is minimal in the presence of fortilin. In addition, BMP4 induces more Msx2 in the absence of fortilin than in its presence. Furthermore, Msx2 overexpression causes MEF to undergo apoptotic cell death. CONCLUSION: We conclude that in early phase of development, fortilin functions as an inhibitor of the BMP pathway. The presence of fortilin in the very early stages of development is required for the survival of embryos. GENERAL SIGNIFICANCE: Abnormalities in the fortilin gene may be associated with early pregnancy loss.


Assuntos
Biomarcadores Tumorais/deficiência , Proteínas Morfogenéticas Ósseas/metabolismo , Perda do Embrião/metabolismo , Transdução de Sinais , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Blastocisto/citologia , Blastocisto/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Cruzamentos Genéticos , Perda do Embrião/patologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Fertilidade , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Saúde , Humanos , Camundongos , Camundongos Mutantes , Neurônios/citologia , Neurônios/metabolismo , Análise de Sobrevida , Proteína Tumoral 1 Controlada por Tradução , Xenopus/embriologia , beta-Galactosidase/metabolismo
18.
Mol Ther ; 17(5): 803-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19277012

RESUMO

Accumulation of aggregated amyloid-beta (Abeta) peptide was studied as an initial step for Alzheimer's disease (AD) pathogenesis. Following amyloid plaque formation, reactive microglia and astrocytes accumulate around plaques and cause neuroinflammation. Here brain chemokines play a major role for the glial accumulation. We have previously shown that transgenic overexpression of chemokine CCL2 in the brain results in increased microglial accumulation and diffuse amyloid plaque deposition in a transgenic mouse model of AD expressing Swedish amyloid precursor protein (APP) mutant. Here, we report that adeno-associated virus (AAV) serotype 1 and 2 hybrid efficiently deliver 7ND gene, a dominant-negative CCL2 mutant, in a dose-response manner and express >1,000-fold higher recombinant CCL2 than basal levels after a single administration. AAV1/2 hybrid virus principally infected neurons without neuroinflammation with sustained expression for 6-months. 7ND expressed in APP/presenilin-1 (APP/PS1) bigenic mice reduced astro/microgliosis, beta-amyloidosis, including suppression of both fibrillar and oligomer Abeta accumulation, and improved spatial learning. Our data support the idea that the AAV1/2 system is a useful tool for CNS gene delivery, and suppression of CCL2 may be a therapeutic target for the amelioration of AD-related neuroinflammation.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Amiloidose/terapia , Quimiocina CCL2/genética , Dependovirus/genética , Gliose/terapia , Aprendizagem em Labirinto/fisiologia , Presenilina-1/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Quimiocina CCL2/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos
19.
J Immunol ; 181(6): 3877-86, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768842

RESUMO

Vaccination therapy of AD animal models and patients strongly suggests an active role of brain mononuclear phagocytes in immune-mediated clearance of amyloid-beta peptides (Abeta) in brain. Although Abeta uptake by macrophages can be regulated by pro- and anti-inflammatory cytokines, their effects on macrophage-mediated Abeta degradation are poorly understood. To better understand this mechanism of degradation, we examined whether pro- and anti-inflammatory cytokines affect the degradation of Abeta using primary cultured human monocyte-derived macrophages (MDM) and microglia using pulse-chase analysis of fibrillar and oligomer (125)I-Abeta40 and Abeta42. Initial uptake of fibrillar Abeta40 and Abeta42 was 40% and its degradation was saturated by 120 h in both MDM and microglia, compared with an initial uptake of oligomeric Abeta less than 0.5% and saturation of degradation within 24 h. IFN-gamma increased the intracellular retention of fibrillar Abeta40 and Abeta42 by inhibiting degradation, whereas IL-4, IL-10, and TGF-beta1, but not IL-13 and IL-27, enhanced degradation. Fibrillar Abeta degradation in MDM is sensitive to lysosomal and insulin degrading enzyme inhibitors but insensitive to proteasomal and neprilysin inhibitors. IFN-gamma and TNF-alpha directly reduced the expression of insulin degrading enzyme and chaperone molecules (heat shock protein 70 and heat shock cognate protein 70), which are involved in refolding of aggregated proteins. Coculture of MDM with activated, but not naive T cells, suppressed Abeta degradation in MDM, which was partially blocked by a combination of neutralizing Abs against proinflammatory cytokines. These data suggest that proinflammatory cytokines suppress Abeta degradation in MDM, whereas select anti-inflammatory and regulatory cytokines antagonize these effects.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Citocinas/fisiologia , Macrófagos/imunologia , Microglia/imunologia , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Acetilação , Peptídeos beta-Amiloides/ultraestrutura , Células Cultivadas , Técnicas de Cocultura , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Radioisótopos do Iodo/metabolismo , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/ultraestrutura , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Microglia/metabolismo , Microglia/ultraestrutura , Microscopia de Força Atômica , Fragmentos de Peptídeos/ultraestrutura
20.
J Neuroimmune Pharmacol ; 2(2): 213-21, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18040846

RESUMO

Accumulating evidence suggests that bone-marrow (BM)-derived mononuclear phagocytes have an important role in the clearance of soluble and aggregated amyloid-beta peptides (Abeta) in Alzheimer's disease (AD) brains. However, the exact kinetics of Abeta clearance in mononuclear phagocytes derived from transgenic animal models of AD expressing beta-amyloid precursor protein (APP) mutants have been poorly characterized. We have examined whether CCL2 and APP expression affects the clearance of Abeta in conjunction with our control, acetylated low-density lipoprotein (AcLDL), using primary cultured BM-derived macrophages derived from adult APP, CCL2, APP/CCL2, and control littermates. Pulse-chase analysis demonstrated three distinct destinations for Abeta40 and AcLDL: intracellular retention, degradation, and secretion. As predicted, 50% of Abeta remained intracellularly contained even 5 days after pulse, while 40% of degraded and 14% of nondegraded Abeta were secreted. APP/CCL2 macrophages show reduced intracellular Abeta retention, along with enhanced secretion of both degraded and nondegraded Abeta. Abeta accumulation in aggresome is also partially reduced in APP/CCL2 macrophages as compared to other APP, CCL2, or control groups, suggesting impaired sorting of aggregated Abeta in aggresomes. The degradation of intracranially injected (125)I-Abeta40 aggregates was also enhanced in adult APP/CCL2 mice as compared to APP littermates in vivo. These data suggest that APP and CCL2 synergistically enhance BM-derived macrophage-mediated clearance of Abeta. In contrast, the clearance of AcLDL by BM-derived macrophages was not significantly enhanced by the presence of either APP or CCL2.


Assuntos
Peptídeos beta-Amiloides/farmacocinética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/farmacocinética , Quimiocina CCL2/genética , Quimiocina CCL2/farmacocinética , Macrófagos/metabolismo , Peptídeos beta-Amiloides/biossíntese , Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células Cultivadas , Quimiocina CCL2/biossíntese , Feminino , Humanos , Macrófagos/patologia , Masculino , Taxa de Depuração Metabólica/genética , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA