Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Acta Neuropathol ; 147(1): 65, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38557897

RESUMO

Human microglia are critically involved in Alzheimer's disease (AD) progression, as shown by genetic and molecular studies. However, their role in tau pathology progression in human brain has not been well described. Here, we characterized 32 human donors along progression of AD pathology, both in time-from early to late pathology-and in space-from entorhinal cortex (EC), inferior temporal gyrus (ITG), prefrontal cortex (PFC) to visual cortex (V2 and V1)-with biochemistry, immunohistochemistry, and single nuclei-RNA-sequencing, profiling a total of 337,512 brain myeloid cells, including microglia. While the majority of microglia are similar across brain regions, we identified a specific subset unique to EC which may contribute to the early tau pathology present in this region. We calculated conversion of microglia subtypes to diseased states and compared conversion patterns to those from AD animal models. Targeting genes implicated in this conversion, or their upstream/downstream pathways, could halt gene programs initiated by early tau progression. We used expression patterns of early tau progression to identify genes whose expression is reversed along spreading of spatial tau pathology (EC > ITG > PFC > V2 > V1) and identified their potential involvement in microglia subtype conversion to a diseased state. This study provides a data resource that builds on our knowledge of myeloid cell contribution to AD by defining the heterogeneity of microglia and brain macrophages during both temporal and regional pathology aspects of AD progression at an unprecedented resolution.


Assuntos
Doença de Alzheimer , Animais , Humanos , Doença de Alzheimer/patologia , Proteínas tau/genética , Proteínas tau/metabolismo , Transcriptoma , Encéfalo/patologia , Células Mieloides/patologia , Microglia/patologia , Peptídeos beta-Amiloides/metabolismo
2.
iScience ; 25(1): 103658, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35072001

RESUMO

Tau pathobiology has emerged as a key component underlying Alzheimer's disease (AD) progression; however, human neuronal in vitro models have struggled to recapitulate tau phenomena observed in vivo. Here, we aimed to define the minimal requirements to achieve endogenous tau aggregation in functional neurons utilizing human induced pluripotent stem cell (hiPSC) technology. Optimized hiPSC-derived cortical neurons seeded with AD brain-derived competent tau species or recombinant tau fibrils displayed increases in insoluble, endogenous tau aggregates. Importantly, MAPT-wild type and MAPT-mutant hiPSC-neurons exhibited unique propensities for aggregation dependent on the seed strain rather than the repeat domain identity, suggesting that successful templating of the recipient tau may be driven by the unique conformation of the seed. The in vitro model presented here represents the first successful demonstration of combining human neurons, endogenous tau expression, and AD brain-derived competent tau species, offering a more physiologically relevant platform to study tau pathobiology.

3.
Cells ; 9(9)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32854315

RESUMO

Pathological hallmarks of Alzheimer's disease (AD) are deposits of amyloid beta (Aß) and hyper-phosphorylated tau aggregates in brain plaques. Recent studies have highlighted the importance of Aß and tau-containing extracellular vesicles (EVs) in AD. We therefore examined EVs separated from cerebrospinal fluid (CSF) of AD, mild cognitive impairment (MCI), and control (CTRL) patient samples to profile the protein composition of CSF EV. EV fractions were separated from AD (n = 13), MCI (n = 10), and CTRL (n = 10) CSF samples using MagCapture Exosome Isolation kit. The CSF-derived EV proteins were identified and quantified by label-free and tandem mass tag (TMT)-labeled mass spectrometry. Label-free proteomics analysis identified 2546 proteins that were significantly enriched for extracellular exosome ontology by Gene Ontology analysis. Canonical Pathway Analysis revealed glia-related signaling. Quantitative proteomics analysis, moreover, showed that EVs expressed 1284 unique proteins in AD, MCI and CTRL groups. Statistical analysis identified three proteins-HSPA1A, NPEPPS, and PTGFRN-involved in AD progression. In addition, the PTGFRN showed a moderate correlation with amyloid plaque (rho = 0.404, p = 0.027) and tangle scores (rho = 0.500, p = 0.005) in AD, MCI and CTRL. Based on the CSF EV proteomics, these data indicate that three proteins, HSPA1A, NPEPPS and PTGFRN, may be used to monitor the progression of MCI to AD.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Doença de Alzheimer/genética , Vesículas Extracelulares/metabolismo , Proteômica/métodos , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Projetos Piloto
4.
Sci Transl Med ; 9(386)2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28424326

RESUMO

Tauopathies are a group of disorders in which the cytosolic protein tau aggregates and accumulates in cells within the brain, resulting in neurodegeneration. A promising treatment being explored for tauopathies is passive immunization with anti-tau antibodies. We previously found that administration of an anti-tau antibody to human tau transgenic mice increased the concentration of plasma tau. We further explored the effects of administering an anti-tau antibody on plasma tau. After peripheral administration of an anti-tau antibody to human patients with tauopathy and to mice expressing human tau in the central nervous system, there was a dose-dependent increase in plasma tau. In mouse plasma, we found that tau had a short half-life of 8 min that increased to more than 3 hours after administration of anti-tau antibody. As tau transgenic mice accumulated insoluble tau in the brain, brain soluble and interstitial fluid tau decreased. Administration of anti-tau antibody to tau transgenic mice that had decreased brain soluble tau and interstitial fluid tau resulted in an increase in plasma tau, but this increase was less than that observed in tau transgenic mice without these brain changes. Tau transgenic mice subjected to acute neuronal injury using 3-nitropropionic acid showed increased interstitial fluid tau and plasma tau. These data suggest that peripheral administration of an anti-tau antibody results in increased plasma tau, which correlates with the concentration of extracellular and soluble tau in the brain.


Assuntos
Anticorpos/farmacologia , Tauopatias/sangue , Tauopatias/metabolismo , Proteínas tau/sangue , Proteínas tau/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Camundongos , Camundongos Transgênicos , Nitrocompostos/toxicidade , Propionatos/toxicidade
5.
Ann Clin Transl Neurol ; 2(3): 278-88, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25815354

RESUMO

OBJECTIVE: We previously found a strong reduction in tau pathology and insoluble tau in P301S tau transgenic mice following intracerebroventricular infusion of the anti-tau antibody HJ8.5. We sought to determine the effects of HJ8.5 in the same model following peripheral administration. METHODS: The primary objective was to determine if HJ8.5 administered at a dose of 50 mg kg(-1) week(-1) by intraperitoneal (IP) injection to 6-month-old P301S mice for 3 months would influence phospho-tau (p-tau) accumulation, tau insolubility, and neurodegeneration. RESULTS: Treatment with HJ8.5 at 50 mg/kg showed a very strong decrease in detergent-insoluble tau. Importantly, HJ8.5 significantly reduced the loss of cortical and hippocampal tissue volumes compared to control treated mice. HJ8.5 treatment reduced hippocampal CA1 cellular layer staining with the p-tau antibody AT8 and thio-S-positive tau aggregates in piriform cortex and amygdala. Moreover, mice treated with HJ8.5 at 50 mg/kg showed a decrease in motor/sensorimotor deficits compared to vehicle-treated mice. Some effects of HJ8.5, including reduction in brain atrophy, and p-tau immunostaining were also seen with a dose of 10 mg kg(-1) week(-1). In BV2-microglial cells, we observed significantly higher uptake of P301S tau aggregates in the presence of HJ8.5. HJ8.5 treatment also resulted in a large dose-dependent increase of tau in the plasma. INTERPRETATION: Our results indicate that systemically administered anti-tau antibody HJ8.5 significantly decreases insoluble tau, decreases brain atrophy, and improves motor/sensorimotor function in a mouse model of tauopathy. These data further support the idea that anti-tau antibodies should be further assessed as a potential treatment for tauopathies.

6.
Neuron ; 80(2): 402-414, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24075978

RESUMO

Tau aggregation occurs in neurodegenerative diseases including Alzheimer's disease and many other disorders collectively termed tauopathies. trans-cellular propagation of tau pathology, mediated by extracellular tau aggregates, may underlie pathogenesis of these conditions. P301S tau transgenic mice express mutant human tau protein and develop progressive tau pathology. Using a cell-based biosensor assay, we screened anti-tau monoclonal antibodies for their ability to block seeding activity present in P301S brain lysates. We infused three effective antibodies or controls into the lateral ventricle of P301S mice for 3 months. The antibodies markedly reduced hyperphosphorylated, aggregated, and insoluble tau. They also blocked development of tau seeding activity detected in brain lysates using the biosensor assay, reduced microglial activation, and improved cognitive deficits. These data imply a central role for extracellular tau aggregates in the development of pathology. They also suggest that immunotherapy specifically designed to block trans-cellular aggregate propagation will be a productive treatment strategy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Encéfalo/metabolismo , Transtornos Cognitivos/tratamento farmacológico , Tauopatias/tratamento farmacológico , Proteínas tau/antagonistas & inibidores , Proteínas tau/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Células Cultivadas , Transtornos Cognitivos/complicações , Transtornos Cognitivos/fisiopatologia , Humanos , Infusões Intraventriculares , Camundongos , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Tauopatias/complicações , Tauopatias/patologia , Tauopatias/psicologia , Proteínas tau/metabolismo , Proteínas tau/toxicidade
7.
Proc Natl Acad Sci U S A ; 110(33): E3138-47, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23898162

RESUMO

Recent experimental evidence suggests that transcellular propagation of fibrillar protein aggregates drives the progression of neurodegenerative diseases in a prion-like manner. This phenomenon is now well described in cell and animal models and involves the release of protein aggregates into the extracellular space. Free aggregates then enter neighboring cells to seed further fibrillization. The mechanism by which aggregated extracellular proteins such as tau and α-synuclein bind and enter cells to trigger intracellular fibril formation is unknown. Prior work indicates that prion protein aggregates bind heparan sulfate proteoglycans (HSPGs) on the cell surface to transmit pathologic processes. Here, we find that tau fibril uptake also occurs via HSPG binding. This is blocked in cultured cells and primary neurons by heparin, chlorate, heparinase, and genetic knockdown of a key HSPG synthetic enzyme, Ext1. Interference with tau binding to HSPGs prevents recombinant tau fibrils from inducing intracellular aggregation and blocks transcellular aggregate propagation. In vivo, a heparin mimetic, F6, blocks neuronal uptake of stereotactically injected tau fibrils. Finally, uptake and seeding by α-synuclein fibrils, but not huntingtin fibrils, occurs by the same mechanism as tau. This work suggests a unifying mechanism of cell uptake and propagation for tauopathy and synucleinopathy.


Assuntos
Amiloide/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Células-Tronco Neurais/metabolismo , Pinocitose/fisiologia , Proteínas tau/metabolismo , Actinas/metabolismo , Animais , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Imuno-Histoquímica , Indóis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Células-Tronco Neurais/fisiologia
8.
Behav Brain Res ; 243: 205-12, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23333843

RESUMO

Animal models of Parkinson's disease (PD) have been widely used to investigate the pathogenesis of this neurodegenerative disorder which is typically associated with the specific and largely disordered protein α-synuclein (α-syn). In the current study, the nasal vector was used to deliver α-syn aggregates to the brain. Both α-syn oligomers and its fibrils were firstly characterized using atomic force microscopy and the thioflavin T binding assay. The toxic oligomers alone (0.48 mg/kg) or their 50:50 combination with fibrils (in a total dose of 0.48 mg/kg) were then given intranasally for ten days in mice and PD-mimetic symptoms as well as humoral immunity to these species and dopamine (DA) were evaluated simultaneously. Open-field behavioral deficits indicated by rigidity and reduced locomotor activity were induced by the dual administration of α-syn oligomers plus fibrils but not the oligomers by themselves under the 10-day dosing regimen. In contrast, using ELISA, high levels of serum autoantibodies to α-syn monomeric, oligomeric and fibrillar conformers as well as DA were observed in both treatment groups reflecting immune system activation and this substantiates previous clinical studies in Parkinson's disease patients. Thus, nasal administration of α-syn amyloidogenic species may be a potential experimental PD model which results not only in motor deficits but also incitement of humoral protection to mimic the disease. Such a paradigm may be exploitable in the quest for potential therapeutic strategies and further studies are warranted.


Assuntos
Comportamento Animal/efeitos dos fármacos , Dopamina/imunologia , Atividade Motora/efeitos dos fármacos , alfa-Sinucleína/farmacologia , Administração Intranasal , Animais , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/imunologia , Rigidez Muscular/imunologia , Doença de Parkinson Secundária/imunologia , Vacinação , alfa-Sinucleína/administração & dosagem
9.
J Biol Chem ; 287(50): 42233-42, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23076148

RESUMO

S100A6 is a small EF-hand calcium- and zinc-binding protein involved in the regulation of cell proliferation and cytoskeletal dynamics. It is overexpressed in neurodegenerative disorders and a proposed marker for Amyotrophic Lateral Sclerosis (ALS). Following recent reports of amyloid formation by S100 proteins, we investigated the aggregation properties of S100A6. Computational analysis using aggregation predictors Waltz and Zyggregator revealed increased propensity within S100A6 helices H(I) and H(IV). Subsequent analysis of Thioflavin-T binding kinetics under acidic conditions elicited a very fast process with no lag phase and extensive formation of aggregates and stacked fibrils as observed by electron microscopy. Ca(2+) exerted an inhibitory effect on the aggregation kinetics, which could be reverted upon chelation. An FT-IR investigation of the early conformational changes occurring under these conditions showed that Ca(2+) promotes anti-parallel ß-sheet conformations that repress fibrillation. At pH 7, Ca(2+) rendered the fibril formation kinetics slower: time-resolved imaging showed that fibril formation is highly suppressed, with aggregates forming instead. In the absence of metals an extensive network of fibrils is formed. S100A6 oligomers, but not fibrils, were found to be cytotoxic, decreasing cell viability by up to 40%. This effect was not observed when the aggregates were formed in the presence of Ca(2+). Interestingly, native S1006 seeds SOD1 aggregation, shortening its nucleation process. This suggests a cross-talk between these two proteins involved in ALS. Overall, these results put forward novel roles for S100 proteins, whose metal-modulated aggregation propensity may be a key aspect in their physiology and function.


Assuntos
Amiloide/química , Cálcio/química , Proteínas de Ciclo Celular/química , Proteínas S100/química , Superóxido Dismutase/química , Tiazóis/química , Amiloide/metabolismo , Amiloide/ultraestrutura , Esclerose Lateral Amiotrófica/metabolismo , Benzotiazóis , Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Humanos , Cinética , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Proteína A6 Ligante de Cálcio S100 , Proteínas S100/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
10.
Neuroimmunomodulation ; 19(6): 334-42, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22986484

RESUMO

OBJECTIVE: Protein aggregation leading to central amyloid deposition is implicated in Parkinson's disease (PD). During disease progression, inflammation and oxidative stress may well invoke humoral immunity against pathological aggregates of PD-associated α-synuclein. The aim was to investigate any possible concurrence between autoimmune responses to α-synuclein monomers, oligomers or fibrils with oxidative stress and inflammation. METHODS: The formation of α-synuclein amyloid species was assessed by thioflavin-T assay and atomic force microscopy was employed to confirm their morphology. Serum autoantibody titers to α-synuclein conformations were determined by ELISA. Enzyme activity and concentrations of oxidative stress/inflammatory indicators were evaluated by enzyme and ELISA protocols. RESULTS: In PD patient sera, a differential increase in autoantibody titers to α-synuclein monomers, toxic oligomers or fibrils was associated with boosted levels of the pro-inflammatory cytokine interleukin-6 and tumour necrosis factor-α, but a decrease in interferon-γ concentration. In addition, levels of malondialdehyde were elevated whilst those of glutathione were reduced along with decrements in the activity of the antioxidants: superoxide dismutase, catalase and glutathione transferase. CONCLUSIONS: It is hypothesized that the generation of α-synuclein amyloid aggregates allied with oxidative stress and inflammatory reactions may invoke humoral immunity protecting against dopaminergic neuronal death. Hence, humoral immunity is a common integrative factor throughout PD progression which is directed towards prevention of further neurodegeneration, so potential treatment strategies should attempt to maintain PD patient immune status.


Assuntos
Autoanticorpos/biossíntese , Estresse Oxidativo/imunologia , Doença de Parkinson/imunologia , Doença de Parkinson/patologia , alfa-Sinucleína/imunologia , Adulto , Idoso , Amiloide/efeitos adversos , Amiloide/imunologia , Amiloide/metabolismo , Autoanticorpos/sangue , Autoanticorpos/uso terapêutico , Feminino , Humanos , Imunidade Humoral/imunologia , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Mediadores da Inflamação/sangue , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Interferon gama/antagonistas & inibidores , Interferon gama/biossíntese , Interferon gama/uso terapêutico , Interleucina-6/biossíntese , Interleucina-6/sangue , Interleucina-6/uso terapêutico , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/prevenção & controle , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/uso terapêutico , alfa-Sinucleína/metabolismo
11.
PLoS One ; 6(4): e18513, 2011 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-21541339

RESUMO

BACKGROUND: Auto-antibodies with specificity to self-antigens have been implicated in a wide variety of neurological diseases, including Parkinson's (PD) and Alzheimer's diseases, being sensitive indicators of neurodegeneration and focus for disease prevention. Of particular interest are the studies focused on the auto-immune responses to amyloidogenic proteins associated with diseases and their applications in therapeutic treatments such as vaccination with amyloid antigens and antibodies in PD, Alzheimer's disease and potentially other neurodegeneration ailments. METHODOLOGY/PRINCIPAL FINDINGS: Generated auto-antibodies towards the major amyloidogenic protein involved in PD Lewy bodies--α-synuclein and its amyloid oligomers and fibrils were measured in the blood sera of early and late PD patients and controls by using ELISA, Western blot and Biacore surface plasmon resonance. We found significantly higher antibody levels towards monomeric α-synuclein in the blood sera of PD patients compared to controls, though the responses decreased with PD progression (P<0.0001). This indicates potential protective role of autoimmunity in maintaining the body homeostasis and clearing protein species whose disbalance may lead to amyloid assembly. There were no noticeable immune responses towards amyloid oligomers, but substantially increased levels of IgGs towards α-synuclein amyloid fibrils both in PD patients and controls, which subsided with the disease progression (P<0.0001). Pooled IgGs from PD patients and controls interacted also with the amyloid fibrils of Aß (1-40) and hen lysozyme, however the latter were recognized with lower affinity. This suggests that IgGs bind to the generic amyloid conformational epitope, displaying higher specificity towards human amyloid species associated with neurodegeneration. CONCLUSIONS/SIGNIFICANCE: Our findings may suggest the protective role of autoimmunity in PD and therefore immune reactions towards PD major amyloid protein--α-synuclein can be of value in the development of treatment and diagnostic strategies, especially during the early disease stages.


Assuntos
Anticorpos/imunologia , Doença de Parkinson/sangue , Doença de Parkinson/diagnóstico , alfa-Sinucleína/sangue , alfa-Sinucleína/imunologia , Amiloide/biossíntese , Amiloide/química , Amiloide/imunologia , Autoimunidade/imunologia , Biomarcadores/sangue , Estudos de Casos e Controles , Reações Cruzadas/imunologia , Feminino , Humanos , Imunoglobulina G/imunologia , Masculino , Pessoa de Meia-Idade , Doença de Parkinson/imunologia , Estrutura Quaternária de Proteína
12.
J Neuroimmunol ; 233(1-2): 221-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21239064

RESUMO

The aim was to ascertain any possible linkage between humoral immune responses to principal biomarkers (α-synuclein monomers, its toxic oligomers or fibrils, dopamine and S100B) and cellular immunity in Parkinson's disease development. There were elevated autoantibody titers to α-synuclein monomers, oligomers plus fibrils in 72%, 56%, and 17% of Parkinsonian patients respectively with a 5-year disease duration. Additionally, there were increased titers to dopamine and S100B (96% and 89%) in the 5-year patient group. All of these values subsided in 10-year sufferers. Furthermore, CD3+, CD4+, CD8+ T-lymphocyte and B-lymphocyte subsets declined in the patient cohort during Parkinsonism indicating disease associated reductions in these lymphocyte subsets.


Assuntos
Doença de Parkinson/imunologia , Doença de Parkinson/metabolismo , Autoanticorpos/biossíntese , Autoanticorpos/sangue , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/patologia , Estudos de Coortes , Progressão da Doença , Dopamina/imunologia , Dopamina/toxicidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Crescimento Neural/imunologia , Fatores de Crescimento Neural/toxicidade , Doença de Parkinson/patologia , Subunidade beta da Proteína Ligante de Cálcio S100 , Proteínas S100/imunologia , Proteínas S100/toxicidade , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , alfa-Sinucleína/imunologia , alfa-Sinucleína/toxicidade
13.
J Mol Biol ; 396(1): 60-74, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-19913026

RESUMO

Amyloid formation is a universal behavior of proteins central to many important human pathologies and industrial processes. The extreme stability of amyloids towards chemical and proteolytic degradation is an acquired property compared to the precursor proteins and is a major prerequisite for their accumulation. Here, we report a study on the lability of human insulin amyloid as a function of pH and amyloid ageing. Using a range of methods such as atomic force microscopy, thioflavin T fluorescence, circular dichroism, and gas-phase electrophoretic mobility macromolecule analysis, we probed the propensity of human insulin amyloid to propagate or dissociate in a wide span of pH values and ageing in a low concentration regime. We generated a three-dimensional amyloid lability landscape in coordinates of pH and amyloid ageing, which displays three distinctive features: (i) a maximum propensity to grow near pH 3.8 and an age corresponding to the inflection point of the growth phase, (ii) an abrupt cutoff between growth and disaggregation at pH 8-10, and (iii) isoclines shifted towards older age during the amyloid growth phase at pH 4-9, reflecting the greater stability of aged amyloid. Thus, lability of amyloid strongly depends on the ionization state of insulin and on the structure and maturity of amyloid fibrils. The stability of insulin amyloid towards protease K was assessed by using real-time atomic force microscopy and thioflavin T fluorescence. We estimated that amyloid fibrils can be digested both from the free ends and within the length of the fibril with a rate of ca 4 nm/min. Our results highlight that amyloid structures, depending on solution conditions, can be less stable than commonly perceived. These results have wide implications for understanding the propagation of amyloids via a seeding mechanism as well as for understanding their natural clearance and dissociation under solution conditions unfavorable for amyloid formation in biological systems and industrial applications.


Assuntos
Amiloide/química , Amiloide/metabolismo , Endopeptidase K/metabolismo , Insulina/química , Insulina/metabolismo , Modelos Moleculares , Amiloide/ultraestrutura , Benzotiazóis , Dicroísmo Circular , Eletroforese , Estabilidade Enzimática , Humanos , Concentração de Íons de Hidrogênio , Cinética , Microscopia de Força Atômica , Ligação Proteica , Estrutura Quaternária de Proteína , Soluções , Análise Espectral , Tiazóis/metabolismo , Fatores de Tempo
14.
PLoS One ; 4(5): e5562, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19440546

RESUMO

BACKGROUND: The conversion of soluble peptides and proteins into polymeric amyloid structures is a hallmark of many age-related degenerative disorders, including Alzheimer's disease, type II diabetes and a variety of systemic amyloidoses. We report here that amyloid formation is linked to another major age-related phenomenon--prostate tissue remodelling in middle-aged and elderly men. METHODOLOGY/PRINCIPAL FINDINGS: By using multidisciplinary analysis of corpora amylacea inclusions in prostate glands of patients diagnosed with prostate cancer we have revealed that their major components are the amyloid forms of S100A8 and S100A9 proteins associated with numerous inflammatory conditions and types of cancer. In prostate protease rich environment the amyloids are stabilized by dystrophic calcification and lateral thickening. We have demonstrated that material closely resembling CA can be produced from S100A8/A9 in vitro under native and acidic conditions and shows the characters of amyloids. This process is facilitated by calcium or zinc, both of which are abundant in ex vivo inclusions. These observations were supported by computational analysis of the S100A8/A9 calcium-dependent aggregation propensity profiles. We found DNA and proteins from Escherichia coli in CA bodies, suggesting that their formation is likely to be associated with bacterial infection. CA inclusions were also accompanied by the activation of macrophages and by an increase in the concentration of S100A8/A9 in the surrounding tissues, indicating inflammatory reactions. CONCLUSIONS/SIGNIFICANCE: These findings, taken together, suggest a link between bacterial infection, inflammation and amyloid deposition of pro-inflammatory proteins S100A8/A9 in the prostate gland, such that a self-perpetuating cycle can be triggered and may increase the risk of malignancy in the ageing prostate. The results provide strong support for the prediction that the generic ability of polypeptide chains to convert into amyloids could lead to their involvement in an increasing number of otherwise apparently unrelated diseases, particularly those associated with ageing.


Assuntos
Envelhecimento , Amiloide/metabolismo , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Neoplasias da Próstata/metabolismo , Idoso , Amiloide/ultraestrutura , Western Blotting , Cálcio/metabolismo , Cromatografia Líquida , DNA Ribossômico/genética , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Microscopia de Força Atômica , Pessoa de Meia-Idade , Análise de Sequência de DNA , Espectrometria de Massas por Ionização por Electrospray , Espectroscopia de Infravermelho com Transformada de Fourier , Zinco/metabolismo
15.
J Mol Biol ; 365(5): 1337-49, 2007 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-17134716

RESUMO

Among the newly discovered amyloid properties, its cytotoxicity plays a key role. Lysozyme is a ubiquitous protein involved in systemic amyloidoses in vivo and forming amyloid under destabilising conditions in vitro. We characterized both oligomers and fibrils of hen lysozyme by atomic force microscopy and demonstrated their dose (5-50 microM) and time-dependent (6-48 h) effect on neuroblastoma SH-SY5Y cell viability. We revealed that fibrils induce a decrease of cell viability after 6 h due to membrane damage shown by inhibition of WST-1 reduction, early lactate dehydrogenase release, and propidium iodide intake; by contrast, oligomers activate caspases after 6 h but cause the cell viability to decline only after 48 h, as shown by fluorescent-labelled annexin V binding to externalized phosphatidylserine, propidium iodide DNA staining, lactate dehydrogenase release, and by typical apoptotic shrinking of cells. We conclude that oligomers induce apoptosis-like cell death, while the fibrils lead to necrosis-like death. As polymorphism is a common property of an amyloid, we demonstrated that it is not a single uniform species but rather a continuum of cross-beta-sheet-containing amyloids that are cytotoxic. An abundance of lysozyme highlights a universal feature of this phenomenon, indicating that amyloid toxicity should be assessed in all clinical applications involving proteinaceous materials.


Assuntos
Amiloide/farmacologia , Apoptose/efeitos dos fármacos , Muramidase/farmacologia , Necrose , Amiloide/isolamento & purificação , Amiloide/ultraestrutura , Animais , Anexina A5/metabolismo , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Eletroforese em Gel de Poliacrilamida , Fluorescência , Interações Hidrofóbicas e Hidrofílicas , Cinética , L-Lactato Desidrogenase/metabolismo , Microscopia de Força Atômica , Muramidase/isolamento & purificação , Muramidase/ultraestrutura , Oxirredução/efeitos dos fármacos , Estrutura Quaternária de Proteína , Solubilidade/efeitos dos fármacos , Sais de Tetrazólio/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...