Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Biol Chem ; 291(20): 10528-40, 2016 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-26957541

RESUMO

The disaccharide trehalose is commonly considered to stimulate autophagy. Cell treatment with trehalose could decrease cytosolic aggregates of potentially pathogenic proteins, including mutant huntingtin, α-synuclein, and phosphorylated tau that are associated with neurodegenerative diseases. Here, we demonstrate that trehalose also alters the metabolism of the Alzheimer disease-related amyloid precursor protein (APP). Cell treatment with trehalose decreased the degradation of full-length APP and its C-terminal fragments. Trehalose also reduced the secretion of the amyloid-ß peptide. Biochemical and cell biological experiments revealed that trehalose alters the subcellular distribution and decreases the degradation of APP C-terminal fragments in endolysosomal compartments. Trehalose also led to strong accumulation of the autophagic marker proteins LC3-II and p62, and decreased the proteolytic activation of the lysosomal hydrolase cathepsin D. The combined data indicate that trehalose decreases the lysosomal metabolism of APP by altering its endocytic vesicular transport.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Trealose/metabolismo , Trealose/farmacologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Células HEK293 , Células Hep G2 , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Fragmentos de Peptídeos/metabolismo , Transporte Proteico/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Serina-Treonina Quinases TOR/metabolismo
2.
Hum Mol Genet ; 23(5): 1365-75, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24154541

RESUMO

Intraneuronal accumulation of ß-amyloid (Aß)42 is one of the earliest pathological events in humans and in animal models of Alzheimer's disease (AD). Apolipoprotein E 4 (APOE4) is the major identified genetic risk factor for late-onset AD, with Aß deposition beginning earlier in apoE4-positive subjects. To directly determine the effects of APOE genotype on intraneuronal accumulation of Aß1-42 at the onset of AD pathogenesis, we introduced lentiviral Aß1-42 into the cortex of APOE targeted replacement (TR) mice at the age of 8-9 months. We demonstrated a significant isoform-dependent effect of human APOE, with dramatically enhanced intracellular Aß1-42 deposits in the cerebral cortex of APOE4-TR mice 2 weeks after injection. Double-immunofluorescent staining showed that intracellular accumulation of lentiviral Aß1-42 was mainly present in neurons, localized to late endosomes/lysosomes. This intraneuronal accumulation of Aß1-42 correlated with increased tau phosphorylation and cell death in the ipsilateral cortex around the injection site. Aß1-42 was also observed in microglia, but not in astrocytes. Quantitative analysis revealed more neurons with Aß1-42 while less microglia with Aß1-42 nearest to the injection site of Aß1-42 lentivirus in APOE4-TR mice. Finally, apoE was present in neurons of the ipsilateral cortex of APOE-TR mice at 2 weeks after lentivirus injection, in addition to astrocytes and microglia in both the ipsilateral and contralateral cerebral cortex. Taken together, these results demonstrate that apoE4 tips the balance of the glial and neuronal Aß toward the intraneuronal accumulation of Aß.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Apolipoproteína E4/genética , Vetores Genéticos/genética , Genótipo , Lentivirus/genética , Neurônios/metabolismo , Transdução Genética , Animais , Apolipoproteína E4/metabolismo , Córtex Cerebral/metabolismo , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Humanos , Espaço Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Microinjeções , Ligação Proteica , Transporte Proteico
3.
J Biol Chem ; 289(24): 16761-72, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24808180

RESUMO

Progressive accumulation of the amyloid ß protein in extracellular plaques is a neuropathological hallmark of Alzheimer disease. Amyloid ß is generated during sequential cleavage of the amyloid precursor protein (APP) by ß- and γ-secretases. In addition to the proteolytic processing by secretases, APP is also metabolized by lysosomal proteases. Here, we show that accumulation of intracellular sphingosine-1-phosphate (S1P) impairs the metabolism of APP. Cells lacking functional S1P-lyase, which degrades intracellular S1P, strongly accumulate full-length APP and its potentially amyloidogenic C-terminal fragments (CTFs) as compared with cells expressing the functional enzyme. By cell biological and biochemical methods, we demonstrate that intracellular inhibition of S1P-lyase impairs the degradation of APP and CTFs in lysosomal compartments and also decreases the activity of γ-secretase. Interestingly, the strong accumulation of APP and CTFs in S1P-lyase-deficient cells was reversed by selective mobilization of Ca(2+) from the endoplasmic reticulum or lysosomes. Intracellular accumulation of S1P also impairs maturation of cathepsin D and degradation of Lamp-2, indicating a general impairment of lysosomal activity. Together, these data demonstrate that S1P-lyase plays a critical role in the regulation of lysosomal activity and the metabolism of APP.


Assuntos
Aldeído Liases/efeitos dos fármacos , Precursor de Proteína beta-Amiloide/metabolismo , Lisossomos/metabolismo , Aldeído Liases/genética , Aldeído Liases/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Cálcio/metabolismo , Catepsina D/metabolismo , Células HEK293 , Humanos , Lisofosfolipídeos/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Camundongos , Proteólise , Esfingosina/análogos & derivados , Esfingosina/metabolismo
4.
Am J Pathol ; 180(6): 2404-16, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22510327

RESUMO

Alzheimer's disease (AD) is the most frequent cause of dementia. There is compelling evidence that the proteolytic processing of the amyloid precursor protein (APP) and accumulation of amyloid-ß (Aß) peptides play critical roles in AD pathogenesis. Due to limited access to human neural tissue, pathogenetic studies have, so far, mostly focused on the heterologous overexpression of mutant human APP in non-human cells. In this study, we show that key steps in proteolytic APP processing are recapitulated in neurons generated from human embryonic and induced pluripotent stem cell-derived neural stem cells (NSC). These human NSC-derived neurons express the neuron-specific APP(695) splice variant, BACE1, and all members of the γ-secretase complex. The human NSC-derived neurons also exhibit a differentiation-dependent increase in Aß secretion and respond to the pharmacotherapeutic modulation by anti-amyloidogenic compounds, such as γ-secretase inhibitors and nonsteroidal anti-inflammatory drugs. Being highly amenable to genetic modification, human NSCs enable the study of mechanisms caused by disease-associated mutations in human neurons. Interestingly, the AD-associated PS1(L166P) variant revealed a partial loss of γ-secretase function, resulting in the decreased production of endogenous Aß40 and an increased Aß42/40 ratio. The PS1(L166P) mutant is also resistant to γ-secretase modulation by nonsteroidal anti-inflammatory drugs. Pluripotent stem cell-derived neurons thus provide experimental access to key steps in AD pathogenesis and can be used to screen pharmaceutical compounds directly in a human neuronal system.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/biossíntese , Mutação , Neurônios/metabolismo , Células-Tronco Pluripotentes/citologia , Presenilina-1/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/fisiologia , Anti-Inflamatórios não Esteroides/farmacologia , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Inibidores Enzimáticos/farmacologia , Humanos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Fragmentos de Peptídeos/biossíntese
5.
J Neurosci ; 31(5): 1837-49, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21289194

RESUMO

Deposition of amyloid ß peptides (Aßs) in extracellular amyloid plaques within the human brain is a hallmark of Alzheimer's disease (AD). Aß derives from proteolytic processing of the amyloid precursor protein (APP) by ß- and γ-secretases. The initial cleavage by ß-secretase results in shedding of the APP ectodomain and generation of APP C-terminal fragments (APP-CTFs), which can then be further processed within the transmembrane domain by γ-secretase, resulting in release of Aß. Here, we demonstrate that accumulation of sphingolipids (SLs), as occurs in lysosomal lipid storage disorders (LSDs), decreases the lysosome-dependent degradation of APP-CTFs and stimulates γ-secretase activity. Together, this results in increased generation of both intracellular and secreted Aß. Notably, primary fibroblasts from patients with different SL storage diseases show strong accumulation of potentially amyloidogenic APP-CTFs. By using biochemical, cell biological, and genetic approaches, we demonstrate that SL accumulation affects autophagic flux and impairs the clearance of APP-CTFs. Thus, accumulation of SLs might not only underlie the pathogenesis of LSDs, but also trigger increased generation of Aß and contribute to neurodegeneration in sporadic AD.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Autofagia , Fibroblastos/metabolismo , Lisossomos/metabolismo , Esfingolipídeos/metabolismo , Animais , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Fibroblastos/ultraestrutura , Imuno-Histoquímica , Lisossomos/genética , Lisossomos/ultraestrutura , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Fragmentos de Peptídeos/metabolismo , Esfingolipídeos/genética , Transfecção
6.
J Biol Chem ; 285(14): 10678-89, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20145244

RESUMO

The beta-amyloid precursor protein (APP) represents a type I transmembrane glycoprotein that is ubiquitously expressed. In the brain, it is a key player in the molecular pathogenesis of Alzheimer disease. Its physiological function is however less well understood. Previous studies showed that APP is up-regulated in prostate, colon, pancreatic tumor, and oral squamous cell carcinoma. In this study, we show that APP has an essential role in growth control of pancreatic and colon cancer. Abundant APP staining was found in human pancreatic adenocarcinoma and colon cancer tissue. Interestingly, treating pancreatic and colon cancer cells with valproic acid (VPA, 2-propylpentanoic acid), a known histone deacetylase (HDAC) inhibitor, leads to up-regulation of GRP78, an endoplasmic reticulum chaperone immunoglobulin-binding protein. GRP78 is involved in APP maturation and inhibition of tumor cell growth by down-regulation of APP and secreted soluble APPalpha. Trichostatin A, a pan-HDAC inhibitor, also lowered APP and increased GRP78 levels. In contrast, treating cells with valpromide, a VPA derivative lacking HDAC inhibitory properties, had no effect on APP levels. VPA did not modify the level of epidermal growth factor receptor, another type I transmembrane protein, and APLP2, a member of the APP family, demonstrating the specificity of the VPA effect on APP. Small interfering RNA-mediated knockdown of APP also resulted in significantly decreased cell growth. Based on these observations, the data suggest that APP down-regulation via HDAC inhibition provides a novel mechanism for pancreatic and colon cancer therapy.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Anticonvulsivantes/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/prevenção & controle , Neoplasias Pancreáticas/prevenção & controle , Receptores de Superfície Celular/metabolismo , Ácido Valproico/farmacologia , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Regulação para Baixo , Chaperona BiP do Retículo Endoplasmático , Inibidores de Histona Desacetilases/farmacologia , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Nexinas de Proteases , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/genética , Células Tumorais Cultivadas
7.
J Biol Chem ; 285(48): 37405-14, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20876579

RESUMO

Epidemiological studies indicate that intake of statins decrease the risk of developing Alzheimer disease. Cellular and in vivo studies suggested that statins might decrease the generation of the amyloid ß-peptide (Aß) from the ß-amyloid precursor protein. Here, we show that statins potently stimulate the degradation of extracellular Aß by microglia. The statin-dependent clearance of extracellular Aß is mainly exerted by insulin-degrading enzyme (IDE) that is secreted in a nonconventional pathway in association with exosomes. Stimulated IDE secretion and Aß degradation were also observed in blood of mice upon peripheral treatment with lovastatin. Importantly, increased IDE secretion upon lovastatin treatment was dependent on protein isoprenylation and up-regulation of exosome secretion by fusion of multivesicular bodies with the plasma membrane. These data demonstrate a novel pathway for the nonconventional secretion of IDE via exosomes. The modulation of this pathway could provide a new strategy to enhance the extracellular clearance of Aß.


Assuntos
Aminoácidos/metabolismo , Peptídeos beta-Amiloides/metabolismo , Exossomos/metabolismo , Espaço Extracelular/metabolismo , Insulisina/metabolismo , Microglia/metabolismo , Regulação para Cima , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Linhagem Celular , Espaço Extracelular/genética , Feminino , Humanos , Insulisina/genética , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico
8.
Curr Res Toxicol ; 2: 19-29, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34345847

RESUMO

A diverse set of environmental contaminants have raised a concern about their potential adverse effects on endocrine signaling. Robust and widely accepted battery of in vitro assays is available to assess the disruption of androgenic and estrogenic pathways. However, such definitive systems to investigate effects on the disruption of thyroid pathways by the xenobiotics are not yet well established. One of the major "Molecular Initiating Events" (MIEs) in thyroid disruption involves targeting of thyroid peroxidase (TPO), a key enzyme involved in thyroid hormone synthesis. TPO catalyzes mono- and diiodination of L-Tyrosine (L-Tyr) to generate 3-Iodo-l-tyrosine (MIT) and 3,5-Diiodo-l-tyrosine (DIT), respectively, followed by the coupling of iodinated tyrosine rings to generate thyroid hormones, 3,3'5-Triiodo-l-thyronine (T3) and Levothyroxine (T4). We sought to develop a robust, sensitive, and rapid in vitro assay systems to evaluate the effects of test chemicals on the multiple catalytic activities of thyroid peroxidase. Simple in vitro assays were designed to study TPO mediated distinct reactions using a single LC-MS/MS method. Herein, we describe a battery of assays to investigate the iodination of L-Tyr to MIT and DIT, MIT to DIT as well as, T3 to T4 catalyzed by rat thyroid TPO. Importantly, two sequential reactions involving mono- and diiodination of L-Tyr could be analyzed in a single assay. The assay that monitors in vitro conversion of DIT to T4 was developed to study the coupling of tyrosine rings. Enzyme kinetics studies revealed distinct characteristics of multiple reactions catalyzed by TPO. Further, the known TPO inhibitors were used to assess their potency towards individual TPO substrates and reactions. The resultant half maximum inhibitory concentration (IC50) values highlighted differential targeting of TPO catalyzed reactions by the same inhibitor. Overall results underscore the need to develop more nuanced approaches that account for distinct multiple catalytic activities of TPO.

9.
J Neurosci ; 28(46): 12097-106, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-19005074

RESUMO

Presenilins (PSs) are components of the gamma-secretase complex that mediates intramembranous cleavage of type I membrane proteins. We show that gamma-secretase is involved in the regulation of cellular lipoprotein uptake. Loss of gamma-secretase function decreased endocytosis of low-density lipoprotein (LDL) receptor. The decreased uptake of lipoproteins led to upregulation of cellular cholesterol biosynthesis by increased expression of CYP51 and enhanced metabolism of lanosterol. Genetic deletion of PS1 or transgenic expression of PS1 mutants that cause early-onset Alzheimer's disease led to accumulation of gamma-secretase substrates and mistargeting of adaptor proteins that regulate endocytosis of the LDL receptor. Consistent with decreased endocytosis of these receptors, PS1 mutant mice have elevated levels of apolipoprotein E in the brain. Thus, these data demonstrate a functional link between two major genetic factors that cause early-onset and late-onset Alzheimer's disease.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Encéfalo/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Lipídeos de Membrana/metabolismo , Receptores de LDL/metabolismo , Animais , Apolipoproteínas E/metabolismo , Endocitose/fisiologia , Feminino , Homeostase/fisiologia , Humanos , Lanosterol/metabolismo , Masculino , Lipídeos de Membrana/genética , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Regulação para Cima/fisiologia
10.
J Neurochem ; 111(3): 669-82, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19712059

RESUMO

Chronic oxidative stress has been causally linked to several neurodegenerative disorders. As sensitivity for oxidative stress greatly differs between brain regions and neuronal cell types, specific cellular mechanisms of adaptation to chronic oxidative stress should exist. Our objective was to identify molecular mechanisms of adaptation of neuronal cells after applying chronic sublethal oxidative stress. We demonstrate that cells resistant to oxidative stress exhibit altered cholesterol and sphingomyelin metabolisms. Stress-resistant cells showed reduced levels of molecules involved in cholesterol trafficking and intracellular accumulation of cholesterol, cholesterol precursors, and metabolites. Moreover, stress-resistant cells exhibited reduced SMase activity. The altered lipid metabolism was associated with enhanced autophagy. Treatment of stress-resistant cells with neutral SMase reversed the stress-resistant phenotype, whereas it could be mimicked by treatment of neuronal cells with a specific inhibitor of neutral SMase. Analysis of hippocampal and cerebellar tissue of mouse brains revealed that the obtained cell culture data reflect the in vivo situation. Stress-resistant cells in vitro showed similar features as the less vulnerable cerebellum in mice, whereas stress-sensitive cells resembled the highly sensitive hippocampal area. These findings suggest an important role of the cell type-specific lipid profile for differential vulnerabilities of different brain areas toward chronic oxidative stress.


Assuntos
Adaptação Fisiológica/fisiologia , Colesterol/metabolismo , Lisossomos/metabolismo , Neurônios/ultraestrutura , Estresse Oxidativo/fisiologia , Esfingomielinas/metabolismo , Adaptação Fisiológica/efeitos dos fármacos , Animais , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Células Clonais , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/citologia , Peróxido de Hidrogênio/farmacologia , Lisossomos/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estatística como Assunto
11.
ALTEX ; 36(3): 373-387, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30865783

RESUMO

Chemical substances that induce an allergic response in skin upon contact are called skin allergens or sensitizers, while chemical substances that elicit an allergic response only in presence of light are called photoallergens or photo sensitizers. The Direct Peptide Reactivity Assay (DPRA, OECD N° 442C, 2015) and the Amino Acid Derivative Reactivity Assay (ADRA) are in chemico assays used to discriminate between allergens and non-allergens. The DPRA and the ADRA, respectively, monitor the depletion of model peptides and modified amino acids induced by crosslinking with test chemicals. In the current study we compared these two assays and analyzed their suitability to predict skin sensitization potential of several chemical substances.  In order to study the combined effect of a chemical compound and UV light, we modified DPRA (photo-DPRA) as well as ADRA (photo-ADRA) by introduction of a photo-irradiation parameter. Analysis using photo-DPRA and photo-ADRA correctly distinguished known photoallergens from non-photoallergens. Upon irradiation, photoallergens selectively showed higher depletion of model peptides or modified amino acids. Thus, photo-DPRA and/or photo-ADRA can serve as non-animal in vitro methods for the identification and assessment of photoallergens/ photosensitizers.


Assuntos
Dermatite Alérgica de Contato/etiologia , Técnicas In Vitro , Transtornos de Fotossensibilidade/etiologia , Pele/química , Raios Ultravioleta/efeitos adversos , Alérgenos/química , Alérgenos/farmacologia , Alternativas aos Testes com Animais , Cromatografia Líquida de Alta Pressão , Humanos , Peptídeos/química
12.
J Neuropathol Exp Neurol ; 67(9): 842-56, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18716559

RESUMO

Apolipoprotein E (apoE) plays a role in the pathogenesis of Alzheimer disease (AD). It is involved in the receptor-mediated cellular clearance of the amyloid beta-protein (Abeta) and in the perivascular drainage of the extracellular fluid. Microvascular changes are also associated with AD and have been discussed as a possible reason for altered perivascular drainage. To further clarify the role of apoE in the perivascular and vascular pathology in AD patients, we studied its occurrence and distribution in the perivascular space, the perivascular neuropil, and in the vessel wall of AD and control cases with and without small vessel disease (SVD). Apolipoprotein E was found in the perivascular space and in the neuropil around arteries of the basal ganglia from control and AD cases disclosing no major differences. Western blot analysis of basal ganglia tissue also revealed no significant differences pertaining to the amount of full-length and C-terminal truncated apoE in AD cases compared with controls. In contrast, Abeta occurred in apoE-positive perivascular astrocytes in AD cases but not in controls. In blood vessels, apoE and immunoglobulin G were detected within the SVD-altered vessel wall. The severity of SVD was associated with the occurrence of apoE in the vessel wall and with that of Abeta in perivascular astrocytes. These results point to an important role of apoE in the perivascular clearance of Abeta in the human brain. The occurrence of apoE and immunoglobulin G in SVD lesions and in the perivascular space suggests that the presence of SVD results in plasma-protein leakage into the brain. It is therefore tempting to speculate that apoE represents a pathogenetic link between SVD and AD.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Apolipoproteínas E/metabolismo , Astrócitos/patologia , Vasos Sanguíneos/patologia , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Astrócitos/metabolismo , Vasos Sanguíneos/metabolismo , Barreira Hematoencefálica/patologia , Western Blotting , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Angiopatia Amiloide Cerebral/complicações , Angiopatia Amiloide Cerebral/patologia , Feminino , Humanos , Imunoglobulina G/metabolismo , Imuno-Histoquímica , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Masculino , Pessoa de Meia-Idade
13.
Front Hum Neurosci ; 9: 231, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25972803

RESUMO

INTRODUCTION: Seven hundred ninety-five thousand Americans will have a stroke this year, and half will have a chronic hemiparesis. Substantial animal literature suggests that the mammalian brain has much potential to recover from acute injury using mechanisms of neuroplasticity, and that these mechanisms can be accessed using training paradigms and neurotransmitter manipulation. However, most of these findings have not been tested or confirmed in the rehabilitation setting, in large part because of the challenges in translating a conceptually straightforward laboratory experiment into a meaningful and rigorous clinical trial in humans. Through presentation of methods for a Phase II trial, we discuss these issues and describe our approach. METHODS: In rodents there is compelling evidence for timing effects in rehabilitation; motor training delivered at certain times after stroke may be more effective than the same training delivered earlier or later, suggesting that there is a critical or sensitive period for strongest rehabilitation training effects. If analogous critical/sensitive periods can be identified after human stroke, then existing clinical resources can be better utilized to promote recovery. The Critical Periods after Stroke Study (CPASS) is a phase II randomized, controlled trial designed to explore whether such a sensitive period exists. We will randomize 64 persons to receive an additional 20 h of upper extremity therapy either immediately upon rehab admission, 2-3 months after stroke onset, 6 months after onset, or to an observation-only control group. The primary outcome measure will be the Action Research Arm Test (ARAT) at 1 year. Blood will be drawn at up to 3 time points for later biomarker studies. CONCLUSION: CPASS is an example of the translation of rodent motor recovery experiments into the clinical setting; data obtained from this single site randomized controlled trial will be used to finalize the design of a Phase III trial.

14.
PLoS One ; 9(3): e93120, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24675880

RESUMO

Under normal conditions, brain apolipoprotein E (apoE) is secreted and lipidated by astrocytes, then taken up by neurons via receptor mediated endocytosis. Free apoE is either degraded in intraneuronal lysosomal compartments or released. Here we identified a novel way by which apoE undergoes proteolysis in the extracellular space via a secreted neuronal protease. We show that apoE is cleaved in neuronal conditioned media by a secreted serine protease. This apoE cleavage was inhibited by PMSF and α1-antichymotrypsin, but not neuroserpin-1 or inhibitors of thrombin and cathepsin G, supporting its identity as a chymotrypsin like protease. In addition, apoE incubation with purified chymotrypsin produced a similar pattern of apoE fragments. Analysis of apoE fragments by mass spectrometry showed cleavages occurring at the C-terminal side of apoE tryptophan residues, further supporting our identification of cleavage by chymotrypsin like protease. Hippocampal neurons were more efficient in mediating this apoE cleavage than cortical neurons. Proteolysis of apoE4 generated higher levels of low molecular weight fragments compared to apoE3. Primary glial cultures released an inhibitor of this proteolytic activity. Together, these studies reveal novel mechanism by which apoE can be regulated and therefore could be useful in designing apoE directed AD therapeutic approaches.


Assuntos
Apolipoproteínas E/metabolismo , Neurônios/metabolismo , Serina Proteases/metabolismo , Sequência de Aminoácidos , Animais , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Apolipoproteínas E/química , Encéfalo/metabolismo , Células Cultivadas , Quimotripsina/metabolismo , Meios de Cultivo Condicionados/química , Espaço Extracelular/metabolismo , Humanos , Dados de Sequência Molecular , Neuroglia/metabolismo , Neuropeptídeos/metabolismo , Proteólise/efeitos dos fármacos , Células Piramidais/metabolismo , Ratos , Serpinas/metabolismo , alfa 1-Antiquimotripsina/metabolismo , Neuroserpina
15.
Autophagy ; 7(6): 645-6, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21460623

RESUMO

Recent work from our laboratory demonstrates that the accumulation of sphingolipids (SLs) decreases the capacity of cells to clear potentially amyloidogenic fragments of the amyloid precursor protein (APP) during autophagy. APP is a type I membrane protein and could undergo sequential proteolytic processing by ß- and γ-secretase resulting in the generation of the amyloid ß-peptide (Aß). Genetic, molecular and biochemical evidence indicates that the accumulation of toxic Aß aggregates plays a critical role in the degeneration of neurons during the pathogenesis of Alzheimer disease (AD). Thus, SL storage could promote the accumulation of Ab in endosomal and lysosomal compartments and thereby induce characteristic cytopathological changes of AD.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Esfingolipídeos/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Autofagia , Membrana Celular/metabolismo , Endossomos/metabolismo , Humanos , Lisossomos/metabolismo , Modelos Biológicos , Neurônios/metabolismo
16.
J Biol Chem ; 284(15): 10138-49, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19176482

RESUMO

Presenilin 1 and 2 (PS) are critical components of the gamma-secretase complex that cleaves type I transmembrane proteins within their transmembrane domains. This process leads to release of proteolytically processed products from cellular membranes and plays an essential role in signal transduction or vital functions as cell adhesion. Here we studied the function of presenilins in cell-matrix interaction of wild-type and PS knock-out mouse embryonic fibroblasts. We found for PS1(-/-) cells an altered morphology with significantly reduced sizes of focal adhesion sites compared with wild type. Cell force analyses on micropatterned elastomer films revealed PS1(-/-) cell forces to be reduced by 50%. Pharmacological inhibition confirmed this function of gamma-secretase in adhesion site and cell force formation. On the regulatory level, PS1 deficiency was associated with strongly decreased phosphotyrosine levels of focal adhesion site-specific proteins. The reduced tyrosine phosphorylation was caused by a down-regulation of c-Src kinase activity primarily at the level of c-Src transcription. The direct regulatory connection between PS1 and c-Src could be identified with ephrinB2 as PS1 target protein. Overexpression of ephrinB2 cytoplasmic domain resulted in its nuclear translocation with increased levels of c-Src and a full complementation of the PS1(-/-) adhesion and phosphorylation phenotype. Cleavage of full-length EB2 and subsequent intracellular domain translocation depended on PS1 as these processes were only found in WT cells. Therefore, we conclude that gamma-secretase is vital for controlling cell adhesion and force formation by transcriptional regulation of c-Src via ephrinB2 cleavage.


Assuntos
Regulação da Expressão Gênica , Presenilina-1/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Citoplasma/metabolismo , Efrina-B2/metabolismo , Fibroblastos/metabolismo , Adesões Focais , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Fenótipo , Processamento de Proteína Pós-Traducional , Tirosina/química
17.
J Biol Chem ; 280(30): 28110-7, 2005 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-15923191

RESUMO

Alzheimer disease is associated with extracellular deposits of amyloid beta-peptides in the brain. Amyloid beta-peptides are generated by proteolytic processing of the beta-amyloid precursor protein by beta- and gamma-secretases. The cleavage by secretases occurs predominantly in post-Golgi secretory and endocytic compartments and is influenced by cholesterol, indicating a role of the membrane lipid composition in proteolytic processing of the beta-amyloid precursor protein. To analyze the role of glycosphingolipids in these processes we inhibited glycosyl ceramide synthase, which catalyzes the first step in glycosphingolipid biosynthesis. The depletion of glycosphingolipids markedly reduced the secretion of endogenous beta-amyloid precursor protein in different cell types, including human neuroblastoma SH-SY5Y cells. Importantly, secretion of amyloid beta-peptides was also strongly decreased by inhibition of glycosphingolipid biosynthesis. Conversely, the addition of exogenous brain gangliosides to cultured cells reversed these effects. Biochemical and cell biological experiments demonstrate that the pharmacological reduction of cellular glycosphingolipid levels inhibited maturation and cell surface transport of the beta-amyloid precursor protein. In the glycosphingolipid-deficient cell line GM95, cellular levels and maturation of beta-amyloid precursor protein were also significantly reduced as compared with normal B16 cells. Together, these data demonstrate that glycosphingolipids are implicated in the regulation of the subcellular transport of the beta-amyloid precursor protein in the secretory pathway and its proteolytic processing. Thus, enzymes involved in glycosphingolipid metabolism might represent targets to inhibit the production of amyloid beta-peptides.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Glicoesfingolipídeos/metabolismo , Doença de Alzheimer/metabolismo , Animais , Transporte Biológico , Biotinilação , Western Blotting , Encéfalo/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Colesterol/metabolismo , Endocitose , Gangliosídeos/metabolismo , Glicoesfingolipídeos/antagonistas & inibidores , Glicoesfingolipídeos/química , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Imunoprecipitação , Metabolismo dos Lipídeos , Melanoma Experimental , Camundongos , Peptídeos/química , Rodaminas/farmacologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA