Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(12): e2111405119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35294277

RESUMO

SignificanceOur results demonstrate the existence of early cellular pathways and network alterations in oligodendrocytes in the alpha-synucleinopathies Parkinson's disease and multiple system atrophy. They further reveal the involvement of an immune component triggered by alpha-synuclein protein, as well as a connection between (epi)genetic changes and immune reactivity in multiple system atrophy. The knowledge generated in this study could be used to devise novel therapeutic approaches to treat synucleinopathies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Atrofia de Múltiplos Sistemas , Doença de Parkinson , Sinucleinopatias , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Atrofia de Múltiplos Sistemas/metabolismo , Oligodendroglia/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
2.
Neuropathol Appl Neurobiol ; 50(1): e12962, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38343067

RESUMO

AIMS: According to Braak's hypothesis, it is plausible that Parkinson's disease (PD) originates in the enteric nervous system (ENS) and spreads to the brain through the vagus nerve. In this work, we studied whether inflammatory bowel diseases (IBDs) in humans can progress with the emergence of pathogenic α-synuclein (α-syn) in the gastrointestinal tract and midbrain dopaminergic neurons. METHODS: We have analysed the gut and the ventral midbrain from subjects previously diagnosed with IBD and form a DSS-based rat model of gut inflammation in terms of α-syn pathology. RESULTS: Our data support the existence of pathogenic α-syn in both the gut and the brain, thus reinforcing the potential role of the ENS as a contributing factor in PD aetiology. Additionally, we have analysed the effect of a DSS-based rat model of gut inflammation to demonstrate (i) the appearance of P-α-syn inclusions in both Auerbach's and Meissner's plexuses (gut), (ii) an increase in α-syn expression in the ventral mesencephalon (brain) and (iii) the degeneration of nigral dopaminergic neurons, which all are considered classical hallmarks in PD. CONCLUSION: These results strongly support the plausibility of Braak's hypothesis and emphasise the significance of peripheral inflammation and the gut-brain axis in initiating α-syn aggregation and transport to the substantia nigra, resulting in neurodegeneration.


Assuntos
Doenças Inflamatórias Intestinais , Doença de Parkinson , Humanos , Ratos , Animais , alfa-Sinucleína/metabolismo , Doença de Parkinson/patologia , Encéfalo/patologia , Inflamação/patologia , Neurônios Dopaminérgicos/metabolismo , Doenças Inflamatórias Intestinais/patologia
3.
Acta Neuropathol ; 146(1): 51-75, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37202527

RESUMO

Parkinson's Disease (PD) is a neurodegenerative and progressive disorder characterised by intracytoplasmic inclusions called Lewy bodies (LB) and degeneration of dopaminergic neurons in the substantia nigra (SN). Aggregated α-synuclein (αSYN) is known to be the main component of the LB. It has also been reported to interact with several proteins and organelles. Galectin-3 (GAL3) is known to have a detrimental function in neurodegenerative diseases. It is a galactose-binding protein without known catalytic activity and is expressed mainly by activated microglial cells in the central nervous system (CNS). GAL3 has been previously found in the outer layer of the LB in post-mortem brains. However, the role of GAL3 in PD is yet to be elucidated. In post-mortem samples, we identified an association between GAL3 and LB in all the PD subjects studied. GAL3 was linked to less αSYN in the LB outer layer and other αSYN deposits, including pale bodies. GAL3 was also associated with disrupted lysosomes. In vitro studies demonstrate that exogenous recombinant Gal3 is internalised by neuronal cell lines and primary neurons where it interacts with endogenous αSyn fibrils. In addition, aggregation experiments show that Gal3 affects spatial propagation and the stability of pre-formed αSyn fibrils resulting in short, amorphous toxic strains. To further investigate these observations in vivo, we take advantage of WT and Gal3KO mice subjected to intranigral injection of adenovirus overexpressing human αSyn as a PD model. In line with our in vitro studies, under these conditions, genetic deletion of GAL3 leads to increased intracellular αSyn accumulation within dopaminergic neurons and remarkably preserved dopaminergic integrity and motor function. Overall, our data suggest a prominent role for GAL3 in the aggregation process of αSYN and LB formation, leading to the production of short species to the detriment of larger strains which triggers neuronal degeneration in a mouse model of PD.


Assuntos
Galectina 3 , Doença de Parkinson , Animais , Humanos , Camundongos , alfa-Sinucleína/metabolismo , Neurônios Dopaminérgicos/metabolismo , Galectina 3/metabolismo , Corpos de Lewy/metabolismo , Doença de Parkinson/metabolismo
4.
J Transl Med ; 20(1): 213, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35562704

RESUMO

This paper is a report of recommendations for addressing translational challenges in amyloid disease research. They were developed during and following an international online workshop organized by the LINXS Institute of Advanced Neutron and X-Ray Science in March 2021. Key suggestions include improving cross-cultural communication between basic science and clinical research, increasing the influence of scientific societies and journals (vis-à-vis funding agencies and pharmaceutical companies), improving the dissemination of negative results, and strengthening the ethos of science.

5.
Nanomedicine ; 43: 102563, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35504462

RESUMO

Light microscopy has been a favorite tool of biological studies for almost a century, recently producing detailed images with exquisite molecular specificity achieving spatial resolution at nanoscale. However, light microscopy is insufficient to provide chemical information as a standalone technique. An increasing amount of evidence demonstrates that optical photothermal infrared microspectroscopy (O-PTIR) is a valuable imaging tool that can extract chemical information to locate molecular structures at submicron resolution. To further investigate the applicability of sub-micron infrared microspectroscopy for biomedical applications, we analyzed the contribution of substrate chemistry to the infrared spectra acquired from individual neurons grown on various imaging substrates. To provide an example of correlative immunofluorescence/O-PTIR imaging, we used immunofluorescence to locate specific organelles for O-PTIR measurement, thus capturing molecular structures at the sub-cellular level directly in cells, which is not possible using traditional infrared microspectroscopy or immunofluorescence microscopy alone.


Assuntos
Espectrofotometria Infravermelho , Microscopia de Fluorescência , Estrutura Molecular , Espectrofotometria Infravermelho/métodos
6.
Int J Mol Sci ; 22(7)2021 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810433

RESUMO

Alzheimer's disease affects millions of lives worldwide. This terminal disease is characterized by the formation of amyloid aggregates, so-called amyloid oligomers. These oligomers are composed of ß-sheet structures, which are believed to be neurotoxic. However, the actual secondary structure that contributes most to neurotoxicity remains unknown. This lack of knowledge is due to the challenging nature of characterizing the secondary structure of amyloids in cells. To overcome this and investigate the molecular changes in proteins directly in cells, we used synchrotron-based infrared microspectroscopy, a label-free and non-destructive technique available for in situ molecular imaging, to detect structural changes in proteins and lipids. Specifically, we evaluated the formation of ß-sheet structures in different monogenic and bigenic cellular models of Alzheimer's disease that we generated for this study. We report on the possibility to discern different amyloid signatures directly in cells using infrared microspectroscopy and demonstrate that bigenic (amyloid-ß, α-synuclein) and (amyloid-ß, Tau) neuron-like cells display changes in ß-sheet load. Altogether, our findings support the notion that different molecular mechanisms of amyloid aggregation, as opposed to a common mechanism, are triggered by the specific cellular environment and, therefore, that various mechanisms lead to the development of Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Amiloide/química , Espectrofotometria Infravermelho/métodos , Doença de Alzheimer/diagnóstico por imagem , Peptídeos beta-Amiloides/metabolismo , Amiloidose/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Camundongos , Microscopia de Fluorescência , Neuroblastoma/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína , Espectroscopia de Infravermelho com Transformada de Fourier , Síncrotrons , alfa-Sinucleína/química
7.
Nanomedicine ; 17: 198-209, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30708052

RESUMO

Poly(propylene imine) dendrimers have been shown to be promising 3-dimensional polymers for the use in the pharmaceutical and biomedical applications. Our aims of this study were first, to synthesize a novel type of dendrimer with poly(propylene imine) core and maltose-histidine shell (G4HisMal) assessing if maltose-histidine shell can improve the biocompatibility and the ability to cross the blood-brain barrier, and second, to investigate the potential of G4HisMal to protect Alzheimer disease transgenic mice from memory impairment. Our data demonstrate that G4HisMal has significantly improved biocompatibility and ability to cross the blood-brain barrier in vivo. Therefore, we suggest that a maltose-histidine shell can be used to improve biocompatibility and ability to cross the blood-brain barrier of dendrimers. Moreover, G4HisMal demonstrated properties for synapse and memory protection when administered to Alzheimer disease transgenic mice. Therefore, G4HisMal can be considered as a promising drug candidate to prevent Alzheimer disease via synapse protection.


Assuntos
Histidina/uso terapêutico , Maltose/uso terapêutico , Transtornos da Memória/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Polipropilenos/uso terapêutico , Doença de Alzheimer/complicações , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/patologia , Animais , Linhagem Celular , Dendrímeros/química , Dendrímeros/farmacocinética , Dendrímeros/uso terapêutico , Histidina/análogos & derivados , Histidina/farmacocinética , Humanos , Maltose/análogos & derivados , Maltose/farmacocinética , Transtornos da Memória/complicações , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Nanopartículas/química , Nanopartículas/uso terapêutico , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/farmacocinética , Polipropilenos/química , Polipropilenos/farmacocinética , Sinapses/efeitos dos fármacos , Sinapses/patologia
8.
Neurobiol Dis ; 113: 1-10, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29414379

RESUMO

Alzheimer's disease (AD) brain tissue can act as a seed to accelerate aggregation of amyloid-ß (Aß) into plaques in AD transgenic mice. Aß seeds have been hypothesized to accelerate plaque formation in a prion-like manner of templated seeding and intercellular propagation. However, the structure(s) and location(s) of the Aß seeds remain unknown. Moreover, in contrast to tau and α-synuclein, an in vitro system with prion-like Aß has not been reported. Here we treat human APP expressing N2a cells with AD transgenic mouse brain extracts to induce inclusions of Aß in a subset of cells. We isolate cells with induced Aß inclusions and using immunocytochemistry, western blot and infrared spectroscopy show that these cells produce oligomeric Aß over multiple replicative generations. Further, we demonstrate that cell lysates of clones with induced oligomeric Aß can induce aggregation in previously untreated N2a APP cells. These data strengthen the case that Aß acts as a prion-like protein, demonstrate that Aß seeds can be intracellular oligomers and for the first time provide a cellular model of nucleated seeding of Aß.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Líquido Intracelular/metabolismo , Placa Amiloide/metabolismo , Proteínas Priônicas/biossíntese , Prosencéfalo/metabolismo , Peptídeos beta-Amiloides/genética , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia , Proteínas Priônicas/genética , Prosencéfalo/patologia
9.
Anal Chem ; 86(24): 12047-54, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25415602

RESUMO

Amyloid peptides are the main component of one of the characteristic pathological hallmarks of Alzheimer's disease (AD): senile plaques. According to the amyloid cascade hypothesis, amyloid peptides may play a central role in the sequence of events that leads to neurodegeneration. However, there are other factors, such as oxidative stress, that may be crucial for the development of the disease. In the present paper, we show that it is possible, by using Fourier tranform infrared (FTIR) microscopy, to co-localize amyloid deposits and lipid peroxidation in tissue slides from patients affected by Alzheimer's disease. Plaques and lipids can be analyzed in the same sample, making use of the characteristic infrared bands for peptide aggregation and lipid oxidation. The results show that, in samples from patients diagnosed with AD, the plaques and their immediate surroundings are always characterized by the presence of oxidized lipids. As for samples from non-AD individuals, those without amyloid plaques show a lower level of lipid oxidation than AD individuals. However, it is known that plaques can be detected in the brains of some non-AD individuals. Our results show that, in such cases, the lipid in the plaques and their surroundings display oxidation levels that are similar to those of tissues with no plaques. These results point to lipid oxidation as a possible key factor in the path that goes from showing the typical neurophatological hallmarks to suffering from dementia. In this process, the oxidative power of the amyloid peptide, possibly in the form of nonfibrillar aggregates, could play a central role.


Assuntos
Doença de Alzheimer/patologia , Imuno-Histoquímica/instrumentação , Imuno-Histoquímica/métodos , Lipídeos/química , Placa Amiloide/química , Espectroscopia de Infravermelho com Transformada de Fourier , Química Encefálica , Humanos , Oxirredução
10.
J Med Chem ; 66(4): 2542-2549, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36599042

RESUMO

Infrared spectroscopic imaging is widely used for the visualization of biomolecule structures, and techniques such as optical photothermal infrared (OPTIR) microspectroscopy can achieve <500 nm spatial resolution. However, these approaches lack specificity for particular cell types and cell components and thus cannot be used as a stand-alone technique to assess their properties. Here, we have developed a novel tool, fluorescently guided optical photothermal infrared microspectroscopy, that simultaneously exploits epifluorescence imaging and OPTIR to perform fluorescently guided IR spectroscopic analysis. This novel approach exceeds the diffraction limit of infrared microscopy and allows structural analysis of specific proteins directly in tissue and single cells. Experiments described herein used epifluorescence to rapidly locate amyloid proteins in tissues or neuronal cultures, thus guiding OPTIR measurements to assess amyloid structures at the subcellular level. We believe that this new approach will be a valuable addition to infrared spectroscopy providing cellular specificity of measurements in complex systems for studies of structurally altered protein aggregates.


Assuntos
Proteínas Amiloidogênicas , Espectrofotometria Infravermelho/métodos , Espectroscopia de Infravermelho com Transformada de Fourier/métodos
11.
Life Sci Alliance ; 6(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37290814

RESUMO

Apolipoprotein E4 (ApoE4) is the most important genetic risk factor for Alzheimer's disease (AD). Among the earliest changes in AD is endosomal enlargement in neurons, which was reported as enhanced in ApoE4 carriers. ApoE is thought to be internalized into endosomes of neurons, whereas ß-amyloid (Aß) accumulates within neuronal endosomes early in AD. However, it remains unknown whether ApoE and Aß intersect intracellularly. We show that internalized astrocytic ApoE localizes mostly to lysosomes in neuroblastoma cells and astrocytes, whereas in neurons, it preferentially localizes to endosomes-autophagosomes of neurites. In AD transgenic neurons, astrocyte-derived ApoE intersects intracellularly with amyloid precursor protein/Aß. Moreover, ApoE4 increases the levels of endogenous and internalized Aß42 in neurons. Taken together, we demonstrate differential localization of ApoE in neurons, astrocytes, and neuron-like cells, and show that internalized ApoE intersects with amyloid precursor protein/Aß in neurons, which may be of considerable relevance to AD.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Humanos , Precursor de Proteína beta-Amiloide/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Peptídeos beta-Amiloides/genética , Doença de Alzheimer/genética , Neurônios/fisiologia
12.
Adv Healthc Mater ; 12(31): e2300987, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37689972

RESUMO

Surgical site infections (SSI) are a clinical and economic burden. Suture-associated SSI may develop when bacteria colonize the suture surface and form biofilms that are resistant to antibiotics. Thrombin-derived C-terminal peptide (TCP)-25 is a host defense peptide with a unique dual mode of action that can target both bacteria and the excessive inflammation induced by bacterial products. The peptide demonstrates therapeutic potential in preclinical in vivo wound infection models. In this study, the authors set out to explore whether TCP-25 can provide a new bioactive innate immune feature to hydrophilic polyglactin sutures (Vicryl). Using a combination of biochemical, biophysical, antibacterial, biofilm, and anti-inflammatory assays in vitro, in silico molecular modeling studies, along with experimental infection and inflammation models in mice, a proof-of-concept that TCP-25 can provide Vicryl sutures with a previously undisclosed host defense capacity, that enables targeting of bacteria, biofilms, and the accompanying inflammatory response, is shown.


Assuntos
Infecções Bacterianas , Poliglactina 910 , Humanos , Camundongos , Animais , Poliglactina 910/uso terapêutico , Suturas , Inflamação/tratamento farmacológico , Infecção da Ferida Cirúrgica/tratamento farmacológico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Infecções Bacterianas/tratamento farmacológico , Peptídeos
13.
Acta Neuropathol Commun ; 11(1): 150, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37715247

RESUMO

The hippocampus is a primary region affected in Alzheimer's disease (AD). Because AD postmortem brain tissue is not available prior to symptomatic stage, we lack understanding of early cellular pathogenic mechanisms. To address this issue, we examined the cellular origin and progression of AD pathogenesis by comparing patient-based model systems including iPSC-derived brain cells transplanted into the mouse brain hippocampus. Proteomic analysis of the graft enabled the identification of pathways and network dysfunction in AD patient brain cells, associated with increased levels of Aß-42 and ß-sheet structures. Interestingly, the host cells surrounding the AD graft also presented alterations in cellular biological pathways. Furthermore, proteomic analysis across human iPSC-based models and human post-mortem hippocampal tissue projected coherent longitudinal cellular changes indicative of early to end stage AD cellular pathogenesis. Our data showcase patient-based models to study the cell autonomous origin and progression of AD pathogenesis.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Animais , Camundongos , Humanos , Proteômica , Autopsia , Hipocampo
14.
Mol Pharm ; 9(3): 458-69, 2012 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-22206488

RESUMO

Alzheimer's disease (AD) is characterized by pathological aggregation of ß-amyloid peptides and MAP-Tau protein. ß-Amyloid (Aß) is a peptide responsible for extracellular Alzheimer's plaque formation. Intracellular MAP-Tau aggregates appear as a result of hyperphosphorylation of this cytoskeletal protein. Small, oligomeric forms of Aß are intermediate products that appear before the amyloid plaques are formed. These forms are believed to be most neurotoxic. Dendrimers are highly branched polymers, which may find an application in regulation of amyloid fibril formation. Several biophysical and biochemical methods, like circular dichroism (CD), fluorescence intensity of thioflavin T and thioflavin S, transmission electron microscopy, spectrofluorimetry (measuring quenching of intrinsic peptide fluorescence) and MTT-cytotoxicity assay, were applied to characterize interactions of cationic phosphorus-containing dendrimers of generation 3 and generation 4 (CPDG3, CPDG4) with the fragment of amyloid peptide (Aß(1-28)) and MAP-Tau protein. We have demonstrated that CPDs are able to affect ß-amyloid and MAP-Tau aggregation processes. A neuro-2a cell line (N2a) was used to test cytotoxicity of formed fibrils and intermediate products during the Aß(1-28) aggregation. It has been shown that CPDs might have a beneficial effect by reducing the system toxicity. Presented results suggest that phosphorus dendrimers may be used in the future as agents regulating the fibrilization processes in Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/química , Dendrímeros/química , Fósforo/química , Proteínas tau/química , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/ultraestrutura , Animais , Linhagem Celular Tumoral , Dicroísmo Circular , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Tirosina/química
15.
Biomacromolecules ; 12(11): 3903-9, 2011 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-21936579

RESUMO

Dendrimers have been proved to interact with amyloids, although most of dendrimers assayed in amyloidogenic systems are toxic to cells. The development of glycodendrimers, poly(propyleneimine) (PPI) dendrimers decorated with maltose (Mal), represents the possibility of using dendrimers with a low intrinsic toxicity. In the present paper we show that fourth (PPI-G4-Mal) and fifth (PPI-G5-Mal) generation glycodendrimers have the capacity to interfere with Alzheimer's amyloid peptide Aß(1-40) fibrilization. The interaction is generation dependent: PPI-G5-Mal blocks amyloid fibril formation generating granular nonfibrillar amorphous aggregates, whereas PPI-G4-Mal generates clumped fibrils at low dendrimer-peptide ratios and amorphous aggregates at high ratios. Both PPI-G4-Mal and PPI-G5-Mal are nontoxic to PC12 and SH-SY5Y cells. PPI-G4-Mal reduces amyloid toxicity by clumping fibrils together, whereas amorphous aggregates are toxic to PC12 cells. The results show that glycodendrimers are promising nontoxic agents in the search for anti-amyloidogenic compounds. Fibril clumping may be an anti-amyloid toxicity strategy.


Assuntos
Peptídeos beta-Amiloides/química , Dendrímeros/química , Maltose/química , Fragmentos de Peptídeos/química , Polipropilenos/química , Doença de Alzheimer/tratamento farmacológico , Amiloide/química , Amiloide/ultraestrutura , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Dendrímeros/farmacologia , Humanos , Cinética , Maltose/farmacologia , Células PC12 , Polipropilenos/farmacologia , Multimerização Proteica , Estrutura Quaternária de Proteína , Ratos
16.
Acta Neuropathol Commun ; 9(1): 153, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34530929

RESUMO

Recent studies suggest that brain cell type specific intracellular environments may play important roles in the generation of structurally different protein aggregates that define neurodegenerative diseases. Using human induced pluripotent stem cells (hiPSC) and biochemical and vibrational spectroscopy techniques, we studied whether Parkinson's disease (PD) patient genomes could modulate alpha-synuclein (aSYN) protein aggregates formation. We found increased ß-sheets and aggregated aSYN in PD patient hiPSC-derived midbrain cells, compared to controls. Importantly, we discovered that aSYN protein aggregation is modulated by patient brain cells' intracellular milieus at the primary nucleation phase. Additionally, we found changes in the formation of aSYN fibrils when employing cellular extracts from familial PD compared to idiopathic PD, in a Thioflavin T-based fluorescence assay. The data suggest that changes in cellular milieu induced by patient genomes trigger structural changes of aSYN potentially leading to the formation of strains having different structures, properties and seeding propensities.


Assuntos
Encéfalo/metabolismo , Líquido Intracelular/metabolismo , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Agregados Proteicos/fisiologia , alfa-Sinucleína/metabolismo , Encéfalo/patologia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Neurônios/patologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , alfa-Sinucleína/genética
17.
Sci Rep ; 11(1): 18368, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526539

RESUMO

Amyloid plaques composed of Aß amyloid peptides and neurofibrillary tangles are a pathological hallmark of Alzheimer Disease. In situ identification of early-stage amyloid aggregates in Alzheimer's disease is relevant for their importance as potential targets for effective drugs. Synchrotron-based infrared imaging is here used to identify early-stage oligomeric/granular aggregated amyloid species in situ in the brain of APP/PS1 transgenic mice for the first time. Also, APP/PS1 mice show fibrillary aggregates at 6 and 12 months. A significant decreased burden of early-stage aggregates and fibrillary aggregates is obtained following treatment with poly(propylene imine) dendrimers with histidine-maltose shell (a neurodegenerative protector) in 6-month-old APP/PS1 mice, thus demonstrating their putative therapeutic properties of in AD models. Identification, localization, and characterization using infrared imaging of these non-fibrillary species in the cerebral cortex at early stages of AD progression in transgenic mice point to their relevance as putative pharmacological targets. No less important, early detection of these structures may be useful in the search for markers for non-invasive diagnostic techniques.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Dendrímeros/uso terapêutico , Polipropilenos/uso terapêutico , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Córtex Cerebral/diagnóstico por imagem , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Dendrímeros/administração & dosagem , Histidina/química , Maltose/química , Camundongos , Camundongos Endogâmicos C57BL , Polipropilenos/administração & dosagem , Espectroscopia de Infravermelho com Transformada de Fourier
18.
Light Sci Appl ; 10(1): 151, 2021 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294676

RESUMO

Alzheimer's disease (AD) is the most common cause of dementia, costing about 1% of the global economy. Failures of clinical trials targeting amyloid-ß protein (Aß), a key trigger of AD, have been explained by drug inefficiency regardless of the mechanisms of amyloid neurotoxicity, which are very difficult to address by available technologies. Here, we combine two imaging modalities that stand at opposite ends of the electromagnetic spectrum, and therefore, can be used as complementary tools to assess structural and chemical information directly in a single neuron. Combining label-free super-resolution microspectroscopy for sub-cellular imaging based on novel optical photothermal infrared (O-PTIR) and synchrotron-based X-ray fluorescence (S-XRF) nano-imaging techniques, we capture elemental distribution and fibrillary forms of amyloid-ß proteins in the same neurons at an unprecedented resolution. Our results reveal that in primary AD-like neurons, iron clusters co-localize with elevated amyloid ß-sheet structures and oxidized lipids. Overall, our O-PTIR/S-XRF results motivate using high-resolution multimodal microspectroscopic approaches to understand the role of molecular structures and trace elements within a single neuronal cell.

19.
Cells ; 10(10)2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34685539

RESUMO

Alzheimer's disease (AD) accounts for about 70% of neurodegenerative diseases and is a cause of cognitive decline and death for one-third of seniors. AD is currently underdiagnosed, and it cannot be effectively prevented. Aggregation of amyloid-ß (Aß) proteins has been linked to the development of AD, and it has been established that, under pathological conditions, Aß proteins undergo structural changes to form ß-sheet structures that are considered neurotoxic. Numerous intensive in vitro studies have provided detailed information about amyloid polymorphs; however, little is known on how amyloid ß-sheet-enriched aggregates can cause neurotoxicity in relevant settings. We used scattering-type scanning near-field optical microscopy (s-SNOM) to study amyloid structures at the nanoscale, in individual neurons. Specifically, we show that in well-validated systems, s-SNOM can detect amyloid ß-sheet structures with nanometer spatial resolution in individual neurons. This is a proof-of-concept study to demonstrate that s-SNOM can be used to detect Aß-sheet structures on cell surfaces at the nanoscale. Furthermore, this study is intended to raise neurobiologists' awareness of the potential of s-SNOM as a tool for analyzing amyloid ß-sheet structures at the nanoscale in neurons without the need for immunolabeling.


Assuntos
Sistemas de Liberação de Fármacos por Nanopartículas/uso terapêutico , Neurônios/fisiologia , Espectrofotometria Infravermelho/métodos , Humanos , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia
20.
Adv Sci (Weinh) ; 7(6): 1903004, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32195099

RESUMO

Loss of memory during Alzheimer's disease (AD), a fatal neurodegenerative disorder, is associated with neuronal loss and the aggregation of amyloid proteins into neurotoxic ß-sheet enriched structures. However, the mechanism of amyloid protein aggregation is still not well understood due to many challenges when studying the endogenous amyloid structures in neurons or in brain tissue. Available methods either require chemical processing of the sample or may affect the amyloid protein structure itself. Therefore, new approaches, which allow studying molecular structures directly in neurons, are urgently needed. A novel approach is tested, based on label-free optical photothermal infrared super-resolution microspectroscopy, to study AD-related amyloid protein aggregation directly in the neuron at sub-micrometer resolution. Using this approach, amyloid protein aggregates are detected at the subcellular level, along the neurites and strikingly, in dendritic spines, which has not been possible until now. Here, a polymorphic nature of amyloid structures that exist in AD transgenic neurons is reported. Based on the findings of this work, it is suggested that structural polymorphism of amyloid proteins that occur already in neurons may trigger different mechanisms of AD progression.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA