Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Cell Physiol ; 239(6): e31218, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38345408

RESUMO

One of the pathological hallmarks of Alzheimer's disease (AD) is the presence of extracellular deposits of amyloid beta (Aß) peptide. In addition to Aß as the core component of the amyloid plaque, the amyloid precursor protein (APP) processing fragment Aß was also found accumulated around the plaque. The APPη pathway, mainly mediated by membrane-type 5 matrix metalloproteinase (MT5-MMP), represents an important factor in AD pathogenesis. The proamyloidogenic features of MT5-MMP could result from interactions with APP when trafficking between organelles, so determination of the location within the cell of APPη cleavage and interacting proteins of MT5-MMP affecting this process will be of priority in understanding the role of MT5-MMP in AD. In the present study, MT5-MMP was found to be located in the nucleus, cytosol, and cytosolic subcellular granules of CHO cells that stably expressed wild-type human APP751. MT5-MMP fusion proteins were constructed that could localize enzyme production in the Golgi apparatus, endosome, ER, mitochondria, or plasma membrane. The fusion proteins significantly increased sAPPη when directed to the endosome, Golgi apparatus, plasma membrane, or mitochondria. Since the C-terminal region of MT5-MMP is responsible for its intracellular location and trafficking, this domain was used as the bait in a yeast two-hybrid screen to identify MT5-MMP protein partners in a human brain cDNA library. Identified binding partners included N4BP2L1, TMX3, EIG121, bridging Integrator 1 (BIN1), RUFY4, HTRA1, and TMEM199. The binding of N4BP2L1, EIG121, BIN1, or TMX3 to MT5-MMP resulted in the most significant increase in sAPPη production. Thus, the action of MT5-MMP on APP occurs in multiple locations within the cell and is facilitated by site-specific binding partners.


Assuntos
Precursor de Proteína beta-Amiloide , Ligação Proteica , Animais , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Membrana Celular/metabolismo , Células CHO , Cricetulus , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Metaloproteinases da Matriz Associadas à Membrana/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Transporte Proteico , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Cricetinae
2.
J Periodontal Res ; 59(3): 512-520, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38243688

RESUMO

BACKGROUND: Periodontitis is a chronic inflammatory disease defined by the pathologic loss of the periodontal ligament and alveolar bone in relation to aging. Although clinical cohort studies reported that periodontitis is significantly elevated in males compared to females, emerging evidence indicates that females with dementia are at a greater risk for periodontitis and decreased alveolar bone. OBJECTIVE: This study aimed to evaluate whether dementia is a potential sex-dependent risk factor for periodontal bone loss using an experimental model of periodontitis induced in the triple transgenic (3x-Tg) dementia-like mice and clinical samples collected from senior 65 plus age patients with diagnosed dementia. MATERIALS AND METHODS: We induced periodontitis in dementia-like triple-transgenic (3x-Tg) male and female mice and age-matched wild-type (WT) control mice by ligature placement. Then, alveolar bone loss and osteoclast activity were evaluated using micro-CT and in situ imaging assays. In addition, we performed dental examinations on patients with diagnosed dementia. Finally, dementia-associated Aß42 and p-Tau (T181) and osteoclastogenic receptor activator of nuclear factor kappa-Β ligand (RANKL) in gingival crevicular fluid (GCF) collected from mice and clinical samples were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS: Alveolar bone loss and in situ osteoclast activity were significantly elevated in periodontal lesions of 3x-Tg females but not males, compared to wild-type control mice. In addition, we also observed that the probing pocket depth (PPD) was also significantly elevated in female patients with dementia. Using ELISA assay, we observed that females had elevated levels of osteoclastogenic RANKL and dementia-associated Aß42 and p-Tau (T181) in the GCF collected from experimental periodontitis lesions and clinical samples. CONCLUSION: Altogether, we demonstrate that females with dementia have an increased risk for periodontal bone loss compared to males.


Assuntos
Perda do Osso Alveolar , Demência , Modelos Animais de Doenças , Camundongos Transgênicos , Periodontite , Ligante RANK , Animais , Feminino , Perda do Osso Alveolar/patologia , Perda do Osso Alveolar/diagnóstico por imagem , Perda do Osso Alveolar/metabolismo , Masculino , Camundongos , Demência/etiologia , Humanos , Idoso , Ligante RANK/análise , Ligante RANK/metabolismo , Fatores Sexuais , Periodontite/complicações , Periodontite/patologia , Microtomografia por Raio-X , Osteoclastos/patologia , Peptídeos beta-Amiloides/metabolismo , Líquido do Sulco Gengival/química , Fragmentos de Peptídeos/análise , Fatores de Risco
3.
J Biol Chem ; 290(14): 9299-309, 2015 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-25713139

RESUMO

Brain accumulation of neurotoxic amyloid ß (Aß) peptide because of increased processing of amyloid precursor protein (APP), resulting in loss of synapses and neurodegeneration, is central to the pathogenesis of Alzheimer disease (AD). Therefore, the identification of molecules that regulate Aß generation and those that cause synaptic damage is crucial for future therapeutic approaches for AD. We demonstrated previously that COPS5 regulates Aß generation in neuronal cell lines in a RanBP9-dependent manner. Consistent with the data from cell lines, even by 6 months, COPS5 overexpression in APΔE9 mice (APΔE9/COPS5-Tg) significantly increased Aß40 levels by 32% (p < 0.01) in the cortex and by 28% (p < 0.01) in the hippocampus, whereas the increases for Aß42 were 37% (p < 0.05) and 34% (p < 0.05), respectively. By 12 months, the increase was even more robust. Aß40 levels increased by 63% (p < 0.001) in the cortex and by 65% (p < 0.001) in the hippocampus. Similarly, Aß42 levels were increased by 69% (p < 0.001) in the cortex and by 71% (p < 0.011) in the hippocampus. Increased Aß levels were translated into an increased amyloid plaque burden both in the cortex (54%, p < 0.01) and hippocampus (64%, p < 0.01). Interestingly, COPS5 overexpression increased RanBP9 levels in the brain, which, in turn, led to increased amyloidogenic processing of APP, as reflected by increased levels of sAPPß and decreased levels of sAPPα. Furthermore, COPS5 overexpression reduced spinophilin in both the cortex (19%, p < 0.05) and the hippocampus (20%, p < 0.05), leading to significant deficits in learning and memory skills. Therefore, like RanBP9, COPS5 also plays a pivotal role in amyloid pathology in vivo.


Assuntos
Encéfalo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Deficiências da Aprendizagem/metabolismo , Transtornos da Memória/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Peptídeo Hidrolases/metabolismo , Placa Amiloide/metabolismo , Animais , Encéfalo/patologia , Complexo do Signalossomo COP9 , Ensaio de Imunoadsorção Enzimática , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Transgênicos , Peptídeo Hidrolases/genética
4.
J Biol Chem ; 288(37): 26668-77, 2013 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-23926111

RESUMO

Increased processing of amyloid precursor protein (APP) and accumulation of neurotoxic amyloid ß peptide (Aß) in the brain is central to the pathogenesis of Alzheimer's disease (AD). Therefore, the identification of molecules that regulate Aß generation is crucial for future therapeutic approaches for AD. We demonstrated previously that RanBP9 regulates Aß generation in a number of cell lines and primary neuronal cultures by forming tripartite protein complexes with APP, low-density lipoprotein-related protein, and BACE1, consequently leading to increased amyloid plaque burden in the brain. RanBP9 is a scaffold protein that exists and functions in multiprotein complexes. To identify other proteins that may bind RanBP9 and regulate Aß levels, we used a two-hybrid analysis against a human brain cDNA library and identified COPS5 as a novel RanBP9-interacting protein. This interaction was confirmed by coimmunoprecipitation experiments in both neuronal and non-neuronal cells and mouse brain. Colocalization of COPS5 and RanBP9 in the same subcellular compartments further supported the interaction of both proteins. Furthermore, like RanBP9, COPS5 robustly increased Aß generation, followed by increased soluble APP-ß (sAPP-ß) and decreased soluble-APP-α (sAPP-α) levels. Most importantly, down-regulation of COPS5 by siRNAs reduced Aß generation, implying that endogenous COPS5 regulates Aß generation. Finally, COPS5 levels were increased significantly in AD brains and APΔE9 transgenic mice, and overexpression of COPS5 strongly increased RanBP9 protein levels by increasing its half-life. Taken together, these results suggest that COPS5 increases Aß generation by increasing RanBP9 levels. Thus, COPS5 is a novel RanBP9-binding protein that increases APP processing and Aß generation by stabilizing RanBP9 protein levels.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Precursor de Proteína beta-Amiloide/fisiologia , Proteínas do Citoesqueleto/fisiologia , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Nucleares/fisiologia , Peptídeo Hidrolases/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Sítios de Ligação , Encéfalo/metabolismo , Complexo do Signalossomo COP9 , Proteínas do Citoesqueleto/metabolismo , Feminino , Biblioteca Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Peptídeo Hidrolases/metabolismo , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Técnicas do Sistema de Duplo-Híbrido
5.
Neurobiol Dis ; 69: 169-79, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24892886

RESUMO

We previously demonstrated that RanBP9 overexpression increased Aß generation and amyloid plaque burden, subsequently leading to robust reductions in the levels of several synaptic proteins as well as deficits in the learning and memory skills in a mouse model of Alzheimer's disease (AD). In the present study, we found striking reduction of spinophilin-immunoreactive puncta (52%, p<0.001) and spinophilin area (62.5%, p<0.001) in the primary cortical neurons derived from RanBP9 transgenic mice (RanBP9-Tg) compared to wild-type (WT) neurons. Similar results were confirmed in WT cortical neurons transfected with EGFP-RanBP9. At 6-months of age, the total spine density in the cortex of RanBP9 single transgenic, APΔE9 double transgenic and APΔE9/RanBP9 triple transgenic mice was similar to WT mice. However, in the hippocampus the spine density was significantly reduced (27%, p<0.05) in the triple transgenic mice compared to WT mice due to reduced number of thin spines (33%, p<0.05) and mushroom spines (22%, p<0.05). This suggests that RanBP9 overexpression in the APΔE9 mice accelerates loss of spines and that the hippocampus is more vulnerable. At 12-months of age, the cortex showed significant reductions in total spine density in the RanBP9 (22%, p<0.05), APΔE9 (19%, p<0.05) and APΔE9/RanBP9 (33%, p<0.01) mice compared to WT controls due to reductions in mushroom and thin spines. Similarly, in the hippocampus the total spine density was reduced in the RanBP9 (23%, p<0.05), APΔE9 (26%, p<0.05) and APΔE9/RanBP9 (39%, p<0.01) mice due to reductions in thin and mushroom spines. Most importantly, RanBP9 overexpression in the APΔE9 mice further exacerbated the reductions in spine density in both the cortex (14%, p<0.05) and the hippocampus (16%, p<0.05). Because dendritic spines are considered physical traces of memory, loss of spines due to RanBP9 provided the physical basis for the learning and memory deficits. Since RanBP9 protein levels are increased in AD brains, RanBP9 might play a crucial role in the loss of spines and synapses in AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/fisiopatologia , Córtex Cerebral/fisiopatologia , Proteínas do Citoesqueleto/metabolismo , Espinhas Dendríticas/fisiologia , Hipocampo/fisiopatologia , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Envelhecimento/patologia , Envelhecimento/fisiologia , Doença de Alzheimer/patologia , Animais , Células Cultivadas , Córtex Cerebral/patologia , Proteínas do Citoesqueleto/genética , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Hipocampo/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/patologia , Neurônios/fisiologia , Proteínas Nucleares/genética , Transfecção
6.
Anal Biochem ; 459: 24-30, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24857774

RESUMO

Activation of nonamyloidogenic processing of amyloid precursor protein (APP) has been hypothesized to be a viable approach for Alzheimer's disease drug discovery. However, until recently, the lack of HTS-compatible assay technologies precluded large scale screening efforts to discover molecules that potentiate nonamyloidogenic pathways. We have developed an HTS-compatible assay based on AlphaLISA technology that quantitatively detects soluble APPα (sAPPα), a marker of nonamyloidogenic processing of APP, released from live cells in low volume, 384-well plates. The assay exhibited good QC parameters (Z'>0.5, S/B>2). A pilot screen of 801 compounds yielded a novel chemotype that increased the release of sAPPα 2-fold at 5µM. These results suggest that the AlphaLISA-based HTS assay is robust and sensitive and can be used to screen large compound collections to discover molecules that potentiate the release of sAPPα. Additionally, we demonstrated that increase of APP processing by nonamyloidogenic pathways will result in decrease of release of amyloidogenic Aß40 fragments.


Assuntos
Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Linhagem Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Humanos , Solubilidade
7.
FASEB J ; 27(12): 4776-89, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23982146

RESUMO

Mitochondrial dysfunction and synaptic damage are important features of Alzheimer's disease (AD) associated with amyloid ß (Aß) and tau. We reported previously that the scaffolding protein RanBP9, which is overall increased in brains of patients with AD and in mutant APP transgenic mice, simultaneously promotes Aß generation and focal adhesion disruption by accelerating the endocytosis of APP and ß1-integrin, respectively. Moreover, RanBP9 induces neurodegeneration in vitro and in vivo and mediates Aß-induced neurotoxicity. Here we show in primary hippocampal neurons that RanBP9 potentiates Aß-induced reactive oxygen species (ROS) overproduction, apoptosis, and calcium deregulation. Analyses of calcium-handling measures demonstrate that RanBP9 selectively delays the clearance of cytosolic Ca(2+) mediated by the mitochondrial calcium uniporter through a process involving the translocation of cofilin into mitochondria and oxidative mechanisms. Further, RanBP9 retards the anterograde axonal transport of mitochondria in primary neurons and decreases synaptic mitochondrial activity in brain. These data indicate that RanBP9, cofilin, and Aß mimic and potentiate each other to produce mitochondrial dysfunction, ROS overproduction, and calcium deregulation, which leads to neurodegenerative changes reminiscent of those seen in AD.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sinalização do Cálcio , Proteínas do Citoesqueleto/metabolismo , Mitocôndrias/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Despolimerização de Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Peptídeos beta-Amiloides/farmacologia , Animais , Apoptose , Transporte Axonal , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Proteínas do Citoesqueleto/genética , Hipocampo/citologia , Potencial da Membrana Mitocondrial , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Nucleares/genética , Fragmentos de Peptídeos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sinapses/metabolismo
8.
Bioorg Med Chem Lett ; 24(18): 4384-4388, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25155386

RESUMO

Alzheimer's disease is a persistent neurodegenerative disorder of elderly characterized clinically by irreversible loss of memory due to accumulation of amyloid beta peptides within the amyloid plaques. We report the parallel synthesis and screening results of diverse substituted di-thiazole piperazine benzamides. A new compound TPI-1917-49 was identified as a promising amyloid reducing agent by lowering the levels of Aß at least in two cell types and in vivo.


Assuntos
Peptídeos beta-Amiloides/antagonistas & inibidores , Benzamidas/farmacologia , Piperazinas/química , Tiazóis/química , Peptídeos beta-Amiloides/metabolismo , Animais , Benzamidas/síntese química , Benzamidas/química , Células CHO , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cricetulus , Relação Dose-Resposta a Droga , Camundongos , Conformação Molecular , Oxirredução/efeitos dos fármacos , Piperazina , Relação Estrutura-Atividade
9.
Front Mol Neurosci ; 17: 1365752, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38476461

RESUMO

The leucine-rich repeat-containing protein 25 (LRRC25) is relatively a novel protein with no information on its role in neuronal or brain function. A recent study suggested LRRC25 is a potential risk factor for Alzheimer's disease (AD). As a first step to understanding LRRC25's role in the brain and AD, we found LRRC25 is expressed in both cell membranes and cytoplasm in a punctuate appearance in astrocytes, microglia, and neurons in cell lines as well as mouse brain. We also found that LRRC25 expression is both age- and brain region-dependent and that 1-day-old (1D) pups expressed the least amount of LRRC25 protein compared to adult ages. In the APΔE9 mice, immunoblot quantified LRRC25 protein levels were increased by 166% (**p < 0.01) in the cortex (CX) and by 215% (***p < 0.001) in the hippocampus (HP) relative to wild-type (WT) controls. Both the brainstem (BS) and cerebellum (CB) showed no significant alterations. In the 3xTg mice, only CX showed an increase of LRRC25 protein by 91% (*p < 0.05) when compared to WT controls although the increased trend was noted in the other brain regions. In the AD patient brains also LRRC25 protein levels were increased by 153% (***p < 0.001) when compared to normal control (NC) subjects. Finally, LRRC25 expression in the iPSC-derived neurons quantified by immunofluorescence was increased by 181% (**p < 0.01) in AD-derived neurons when compared to NC-derived neurons. Thus increased LRRC25 protein in multiple models of AD suggests that LRRC25 may play a pathogenic role in either Aß or tau pathology in AD. The mechanism for the increased levels of LRRC25 in AD is unknown at present, but a previous study showed that LRRC25 levels also increase during neonatal hypoxic-ischemia neuronal damage. Based on the evidence that autophagy is highly dysregulated in AD, the increased LRRC25 levels may be due to decreased autophagic degradation of LRRC25. Increased LRRC25 in turn may regulate the stability or activity of key enzymes involved in either Aß or hyperphosphorylated tau generation and thus may contribute to increased plaques and neurofibrillary tangles.

10.
BMC Med ; 11: 81, 2013 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-23531149

RESUMO

BACKGROUND: Currently available therapies for Alzheimer's disease (AD) do not treat the underlying cause of AD. Anecdotal observations in nursing homes from multiple studies strongly suggest an inverse relationship between cancer and AD. Therefore, we reasoned that oncology drugs may be effective against AD. METHODS: We screened a library of all the FDA-approved oncology drugs and identified bis-chloroethylnitrosourea (BCNU or carmustine) as an effective amyloid beta (Aß) reducing compound. To quantify Aß levels, Chinese hamster ovary (CHO) cells stably expressing amyloid precursor protein 751WT (APP751WT) called 7WD10 cells were exposed to different concentrations of BCNU for 48 hours and the conditioned media were collected. To detect Aß the conditioned media were immunoprecipitated with Ab9 antibody and subjected to immunoblot detection. Amyloid plaques were quantified in the brains of a mouse model of AD after chronic exposure to BCNU by thoflavin S staining. RESULTS: BCNU decreased normalized levels of Aß starting from 5 µM by 39% (P < 0.05), 10 µM by 51% (P < 0.01) and 20 µM by 63% (P < 0.01) in CHO cells compared to a control group treated with butyl amine, a structural derivative of BCNU. Interestingly, soluble amyloid precursor protein α (sAPPα) levels were increased to 167% (P < 0.01) at 0.5 µM, 186% (P < 0.05) at 1 µM, 204% (P < 0.01) at 5 µM and 152% (P < 0.05) at 10 µM compared to untreated cells. We also tested the effects of 12 structural derivatives of BCNU on Aß levels, but none of them were as potent as BCNU. BCNU treatment at 5 µM led to an accumulation of immature APP at the cell surface resulting in an increased ratio of surface to total APP by 184% for immature APP, but no change in mature APP. It is also remarkable that BCNU reduced Aß generation independent of secretases which were not altered up to 40 µM. Interestingly, levels of transforming growth factor beta (TGFß) were increased at 5 µM (43%, P < 0.05), 10 µM (73%, P < 0.01) and 20 µM (92%, P < 0.001). Most significantly, cell culture results were confirmed in vivo after chronic administration of BCNU at 0.5 mg/kg which led to the reduction of Aß40 by 75% and amyloid plaque burden by 81%. Conversely, the levels of sAPPα were increased by 45%. CONCLUSIONS: BCNU reduces Aß generation and plaque burden at non-toxic concentrations possibly through altered intracellular trafficking and processing of APP. Taken together these data provided unequivocal evidence that BCNU is a potent secretase-sparing anti-Aß drug. See related commentary article here http://www.biomedcentral.com/1741-7015/11/82.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Antineoplásicos Alquilantes/administração & dosagem , Carmustina/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Placa Amiloide/patologia , Animais , Encéfalo/patologia , Células CHO , Cricetinae , Cricetulus , Modelos Animais de Doenças , Camundongos , Resultado do Tratamento
11.
FASEB J ; 26(4): 1672-81, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22223749

RESUMO

Accumulation of the amyloid ß (Aß) peptide derived from the amyloid precursor protein (APP) plays a central role in the pathogenesis of Alzheimer's disease (AD). We previously reported that the scaffolding protein RanBP9 is markedly increased in AD brains and promotes Aß generation by scaffolding APP/BACE1/LRP complexes together and accelerating APP endocytosis. Because APP, LRP, and RanBP9 all physically interact with ß-integrins, we investigated whether RanBP9 alters integrin-dependent cell adhesion and focal adhesion signaling. Here, we show that RanBP9 overexpression dramatically disrupts integrin-dependent cell attachment and spreading in NIH3T3 and hippocampus-derived HT22 cells, concomitant with strongly decreased Pyk2/paxillin signaling and talin/vinculin localization in focal adhesion complexes. Conversely, RanBP9 knockdown robustly promotes cell attachment, spreading, and focal adhesion signaling and assembly. Cell surface biotinylation and endocytosis assays reveal that RanBP9 overexpression and RanBP9 siRNA potently reduces and increases surface ß1-integrin and LRP by accelerating and inhibiting their endocytosis, respectively. Primary hippocampal neurons derived from RanBP9-transgenic mice also demonstrate severely reduced levels of surface ß1-integrin, LRP, and APP, as well as neurite arborization. Therefore, these data indicate that RanBP9 simultaneously inhibits cell-adhesive processes and enhances Aß generation by accelerating APP, LRP, and ß1-integrin endocytosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Adesões Focais/metabolismo , Integrina beta1/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Adesão Celular/fisiologia , Células Cultivadas , Proteínas do Citoesqueleto/genética , Endocitose/fisiologia , Hipocampo/citologia , Humanos , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas Relacionadas a Receptor de LDL/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/fisiologia , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Talina/genética , Talina/metabolismo , Vinculina/genética , Vinculina/metabolismo
12.
FASEB J ; 26(5): 2072-83, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22294787

RESUMO

We previously reported that RanBP9 binds low-density lipoprotein receptor-related protein (LRP), amyloid precursor protein (APP), and BACE1 and robustly increased Aß generation in a variety of cell lines and primary neuronal cultures. To confirm the physiological/ pathological significance of this phenotype in vivo, we successfully generated transgenic mice overexpressing RanBP9 as well as RanBP9-null mice. Here we show that RanBP9 overexpression resulted in >2-fold increase in Aß40 levels as early as 4 mo of age. A sustained increase in Aß40 levels was seen at 12 mo of age in both CHAPS-soluble and formic acid (FA)-soluble brain fractions. In addition, Aß42 levels were also significantly increased in FA-soluble fractions at 12 mo of age. More important, increased Aß levels were translated to increased deposition of amyloid plaques. In addition, RanBP9 overexpression significantly decreased the levels of synaptophysin and PSD-95 proteins. Conversely, RanBP9-null mice showed increased levels of synaptophysin, PSD-95, and drebrin A protein levels. Given that loss of synapses is the best pathological correlate of cognitive deficits in Alzheimer's disease (AD), increased Aß levels by RanBP9 observed in the present study provides compelling evidence that RanBP9 may indeed play a key role in the etiology of AD. If so, RanBP9 provides a great opportunity to develop novel therapy for AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas do Citoesqueleto/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/fisiologia , Sinapses/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas do Citoesqueleto/genética , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/genética
13.
Front Cell Neurosci ; 16: 954071, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35928571

RESUMO

Alzheimer's disease (AD) is complex and highly heterogeneous. Less than 10% of AD cases are early-onset (EOAD) caused by autosomal dominantly inherited mutations in amyloid precursor protein (APP), presenilin 1 (PS1), or presenilin 2 (PS2), each of which can increase Aß generation and, thus, amyloid plaques. The remaining 90% of cases of AD are late-onset (LOAD) or sporadic. Intense research efforts have led to identification of many genes that increase the risk of AD. An IQ motif containing protein K (IQCK) was recently identified by several investigators as an Alzheimer's disease risk gene. However, how IQCK increases AD risk is completely unknown. Since IQCK is a novel gene, there is limited information on its physiological characterization. To understand its role in AD, it is first important to determine its subcellular localization, whether and where it is expressed in the brain, and what type of brain cells express the IQCK protein. Therefore, in this study, we show by immunocytochemical (ICC) staining that IQCK is expressed in both the nucleus and the cytoplasm of SH-SY5Y neuroblastoma cells as well as HeLa cells but not in either HMC3 microglial or CHO cells. By immunohistochemistry (IHC), we also show that IQCK is expressed in both mouse and human neurons, including neuronal processes in vivo in the mouse brain. IHC data also show that the IQCK protein is widely expressed throughout the mouse brain, although regional differences were noted. IQCK expression was highest in the brainstem (BS), followed by the cerebellum (CB) and the cortex (CX), and it was lowest in the hippocampus (HP). This finding was consistent with data from an immunoblot analysis of brain tissue homogenates. Interestingly, we found IQCK expression in neurons, astrocytes, and oligodendrocytes using cell-specific antibodies, but IQCK was not detected in microglial cells, consistent with negative in vitro results in HMC3 cells. Most importantly, we found that actin-normalized IQCK protein levels were increased by 2 folds in AD brains relative to normal control (NC) brains. Furthermore, the IQCK protein was found in amyloid plaques, suggesting that IQCK may play a pathogenic role in either Aß generation or amyloid plaque deposition in AD.

14.
FASEB J ; 24(1): 119-27, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19729516

RESUMO

Increasing biochemical and genetic evidence indicates that the amyloid-beta (Abeta) peptide derived from amyloid precursor protein (APP) plays a central role in Alzheimer's disease (AD) pathogenesis. We previously reported that RanBP9 promotes Abeta generation by scaffolding APP/BACE1/LRP complexes together. Interestingly, the RanBP9-Delta1/N60 (residues 1-392) deletion mutant interacted much more strongly with APP/BACE1/LRP than full-length RanBP9. In this study, we found that RanBP9-N60, a processed form of RanBP9 virtually identical to the RanBP9-Delta1/N60 mutant, was strongly increased in AD brains compared with controls. To evaluate the potential pathogenic consequences of this phenotype, we studied the differential biological properties of full-length RanBP9 vs. RanBP9-Delta1/N60 in HEK293T and Neuro-2A cells. The RanBP9-Delta1/N60 fragment, which lacks a nuclear localization signal, displayed enhanced cytoplasmic vs. nuclear localization and >3-fold enhanced stability than full-length RanBP9. Importantly, RanBP9-Delta1/N60, which contains the LisH dimerization domain, retained the capacity to form self-interacting multimeric complexes and increased Abeta generation by approximately 5-fold over vector controls, more potent than the approximately 3-fold increase seen by full-length RanBP9. Taken together, these data indicate that RanBP9-N60 may further drive the amyloid cascade in AD and that the proteolytic processing of RanBP9 may be an attractive therapeutic target.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Encéfalo/metabolismo , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células CHO , Estudos de Casos e Controles , Linhagem Celular , Cricetinae , Cricetulus , Proteínas do Citoesqueleto/genética , Humanos , Técnicas In Vitro , Complexos Multiproteicos , Sinais de Localização Nuclear/química , Sinais de Localização Nuclear/genética , Proteínas Nucleares/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência
15.
Mol Neurobiol ; 58(5): 2269-2283, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33417226

RESUMO

Nonamyloidogenic processing of amyloid precursor protein (APP) by augmenting ADAM10 is a promising therapeutic strategy for Alzheimer's disease (AD). Therefore identification of molecular pathways that regulate ADAM10 expression is crucial. Autophagy is strongly dysregulated in AD, and TFEB was recently shown to be a master regulator of autophagy-lysosome pathway (ALP). Here, we report that TFEB expression in HeLa cells increased ADAM10 mature form by 72% (p < 0.01, n = 4), while TFEB knockdown by CRISPR strategy reduced ADAM10 mature form by 36% (p < 0.05, n = 4). Autophagy inhibition by 3-methyladenine (3-MA), but not bafilomycin A1 (BAF1), reduced ADAM10 mature form by 49% (p < 0.05, n = 4) in the TFEB expressing HeLa cells. Autophagy activation by 3 h of starvation increased ADAM10 to 91% (p < 0.001, n = 6) relative to 51% (p < 0.01, n = 6) in the nutrient-fed cells. Further, siRNAs targeted against PPARα in HeLa cells decreased ADAM10 levels by 28% (p < 0.05, n = 6) relative to the cells treated with scrambled siRNAs. Further, incubation of EGFP-TFEB expressing HeLa cells with PPARα antagonist, but not PPARß or PPARγ antagonists, prevented TFEB-induced increase in ADAM10 levels. Importantly, flag-TFEB expression in the brain also increased ADAM10 by 60% (p < 0.05, n = 3) in the cortical and 34% (p < 0.001, n = 3) in the hippocampal homogenates. ADAM10 activity also increased by 57% (p < 0.01, n = 3) in the HeLa cells. Finally, TFEB-induced ADAM10 potentiation led to increased secretion of sAPPα by 154% (p < 0.001, n = 3) in the cortex and 62% (p < 0.001, n = 3) in the hippocampus. Thus, TFEB expression enhances nonamyloidogenic processing of APP. In conclusion, TFEB expression induces ADAM10 in an autophagy-dependent manner through PPARα.


Assuntos
Proteína ADAM10/metabolismo , Autofagia/fisiologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Encéfalo/metabolismo , Neurônios/metabolismo , PPAR alfa/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células HeLa , Humanos , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Sinaptossomos/metabolismo
16.
Mol Neurobiol ; 58(6): 2465-2480, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33439437

RESUMO

The recent outbreak of SARS-CoV-2 infections that causes coronavirus-induced disease of 2019 (COVID-19) is the defining and unprecedented global health crisis of our time in both the scale and magnitude. Although the respiratory tract is the primary target of SARS-CoV-2, accumulating evidence suggests that the virus may also invade both the central nervous system (CNS) and the peripheral nervous system (PNS) leading to numerous neurological issues including some serious complications such as seizures, encephalitis, and loss of consciousness. Here, we present a comprehensive review of the currently known role of SARS-CoV-2 and identify all the neurological problems reported among the COVID-19 case reports throughout the world. The virus might gain entry into the CNS either through the trans-synaptic route via the olfactory neurons or through the damaged endothelium in the brain microvasculature using the ACE2 receptor potentiated by neuropilin-1 (NRP-1). The most critical of all symptoms appear to be the spontaneous loss of breathing in some COVID-19 patients. This might be indicative of a dysfunction within the cardiopulmonary regulatory centers in the brainstem. These pioneering studies, thus, lay a strong foundation for more in-depth basic and clinical research required to confirm the role of SARS-CoV-2 infection in neurodegeneration of critical brain regulatory centers.


Assuntos
COVID-19/complicações , Doenças do Sistema Nervoso Central/etiologia , Doenças do Sistema Nervoso Periférico/etiologia , SARS-CoV-2 , Adulto , Fatores Etários , Enzima de Conversão de Angiotensina 2/metabolismo , Encéfalo/virologia , COVID-19/epidemiologia , COVID-19/fisiopatologia , Doenças Cardiovasculares/epidemiologia , Doenças do Sistema Nervoso Central/diagnóstico por imagem , Doenças do Sistema Nervoso Central/fisiopatologia , Criança , Comorbidade , Diabetes Mellitus/epidemiologia , Células Endoteliais/patologia , Feminino , Humanos , Nefropatias/etiologia , Hepatopatias/etiologia , Masculino , Proteínas do Tecido Nervoso/metabolismo , Neuroimagem , Neuropilina-1/fisiologia , Obesidade/epidemiologia , Especificidade de Órgãos , Doenças do Sistema Nervoso Periférico/fisiopatologia , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo
17.
Front Immunol ; 11: 591571, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329577

RESUMO

Background: Among different types of sphingolipids produced by human cells, the possible engagement of ceramide species in the pathogenesis of Alzheimer's disease (AD) has attracted recent attention. While ceramides are primarily generated by de novo synthesis in mammalian cells, only a limited number of bacterial species, produce ceramides, including phosphoglycerol dihydroceramide (PGDHC) that is produced by the key periodontal pathogen Porphyromonas gingivalis. Emerging evidence indicates that virulence factors produced by P. gingivalis, such as lipopolysaccharide and gingipain, may be engaged in the initiation and/or progression of AD. However, the potential role of PGDHC in the pathogenesis of AD remains unknown. Therefore, the aim of this study was to evaluate the influence of PGDHC on hallmark findings in AD. Material and Methods: CHO-7WD10 and SH-SY-5Y cells were exposed to PGDHC and lipopolysaccharide (LPS) isolated from P. gingivalis. Soluble Aß42 peptide, amyloid precursor protein (APP), phosphorylated tau and senescence-associated secretory phenotype (SASP) factors were quantified using ELISA and Western blot assays. Results: Our results indicate that P. gingivalis (Pg)-derived PGDHC, but not Pg-LPS, upregulated secretion of soluble Aß42 peptide and expression of APP in CHO-7WD10 cells. Furthermore, hyperphosphorylation of tau protein was observed in SH-SY-5Y cells in response to PGDHC lipid. In contrast, Pg-LPS had little, or no significant effect on the tau phosphorylation induced in SH-SY-5Y cells. However, both PGDHC and Pg-LPS contributed to the senescence of SH-SY5Y cells as indicated by the production of senescence-associated secretory phenotype (SASP) markers, including beta-galactosidase, cathepsin B (CtsB), and pro-inflammatory cytokines TNF-α, and IL-6. Additionally, PGDHC diminished expression of the senescence-protection marker sirtuin-1 in SH-SY-5Y cells. Conclusions: Altogether, our results indicate that P. gingivalis-derived PGDHC ceramide promotes amyloidogenesis and hyperphosphorylation, as well as the production of SASP factors. Thus, PGDHC may represent a novel class of bacterial-derived virulence factors for AD associated with periodontitis.


Assuntos
Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Ceramidas/biossíntese , Suscetibilidade a Doenças , Periodontite/complicações , Periodontite/microbiologia , Porphyromonas gingivalis/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Linhagem Celular , Humanos , Fosforilação , Proteínas tau/metabolismo
18.
PLoS One ; 14(4): e0215713, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30995273

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0085484.].

19.
J Control Release ; 314: 125-140, 2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-31647979

RESUMO

Alzheimer's disease (AD) is an irreversible and progressive neurodegenerative disorder manifested by memory loss and cognitive impairment. Deposition of the amyloid ß plaques has been identified as the most common AD pathology; however, the excessive accumulation of phosphorylated or total tau proteins, reactive oxygen species, and higher acetylcholinesterase activity are also strongly associated with Alzheimer's dementia. Several therapeutic approaches targeting these pathogenic mechanisms have failed in clinical or preclinical trials, partly due to the limited bioavailability, poor cell, and blood-brain barrier penetration, and low drug half-life of current regimens. The nanoparticles (NPs)-mediated drug delivery systems improve drug solubility and bioavailability, thus renders as superior alternatives. Moreover, NPs-mediated approaches facilitate multiple drug loading and targeted drug delivery, thereby increasing drug efficacy. However, certain NPs can cause acute toxicity damaging cellular and tissue architecture, therefore, NP material should be carefully selected. In this review, we summarize the recent NPs-mediated studies that exploit various pathologic mechanisms of AD by labeling, identifying, and treating the affected brain pathologies. The disadvantages of the select NP-based deliveries and the future aspects will also be discussed.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Nanopartículas , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/fisiopatologia , Peptídeos beta-Amiloides/metabolismo , Animais , Disponibilidade Biológica , Barreira Hematoencefálica/metabolismo , Humanos , Placa Amiloide , Solubilidade
20.
Neuroscience ; 402: 11-22, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30677488

RESUMO

Abnormalities of the autophagy-lysosomal pathway (ALP) have been implicated in the pathology of Alzheimer's disease (AD). Activation of TFEB (transcription factor EB), a master regulator of the ALP, leads to ALP facilitation. The present study sought to clarify whether TFEB-mediated ALP facilitation influences the process of amyloid ß-protein (Aß) generation in neurons. TFEB was overexpressed in mature rat primary cortical neurons via recombinant adenoviruses, without (basal conditions) or with co-overexpression of wild-type amyloid precursor protein (APP) or its ß-C-terminal fragment (ß-CTF). We confirmed that TFEB overexpression upregulated the lysosomal proteins, cathepsin D and LAMP-1. In TFEB-expressing neurons, protein levels of ADAM10 were profoundly increased, whereas those of APP, BACE1, or γ-secretase complex proteins were unaffected. However, TFEB did not affect ADAM10 mRNA levels. TFEB overexpression had different effects on Aß production depending on the expression level of APP or ß-CTF: TFEB slightly decreased Aß secretion under basal conditions; clearly increased α-CTF levels and marginally increased ß-CTF levels with modest increases in secreted Aß in APP-expressing neurons; and caused a remarkable increase in ß-CTF levels with a significant increase in secreted Aß in ß-CTF-expressing neurons. Inhibition of proteasomes, but not lysosomes, markedly increased ß-CTF levels in ß-CTF-expressing neurons. These results collectively indicate that TFEB modulates Aß production not only by increasing α-secretase processing of APP through ADAM10 upregulation but also by augmenting ß-CTF levels possibly via altered proteasome-mediated catabolism. Thus, TFEB-mediated ALP enhancement appears to have dual, but opposite, effects on Aß production in neurons.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Córtex Cerebral/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Proteína ADAM10/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Fragmentos de Peptídeos/metabolismo , Cultura Primária de Células , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos Wistar , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa