Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Dis ; 163: 105599, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34952161

RESUMO

Alpha-synuclein aggregates are the hallmark pathology of Parkinson's disease, which can propagate in a stereotypical pattern along the brain-body axis. Parkinson's disease patients not only display heterogeneous symptoms but also show variable patterns of alpha-synuclein pathology and affected neuronal systems during the disease course, complicating early and accurate diagnosis. Emerging data from post-mortem and imaging studies strongly suggest that disease heterogeneity could, at least in part, be explained by variable disease onset site, i.e. brain or body. This has led to the recently hypothesized formulation of two Parkinson's disease-subtypes, a body-first subtype where pathogenic alpha-synuclein arises in the body and spreads to the brain, and a brain-first subtype where pathogenic alpha-synuclein arises in the brain and spreads to the body. From a preclinical perspective, several animal models have been adapted or developed to reproduce Parkinson's disease-like pathology in the brain or periphery aiming to address the site of disease onset. Here, we review the current rodent and primate models that aim to reproduce Parkinson's disease pathology development and spreading in the brain and/or body and discuss the value and shortcomings of these models for the development of potential future applications in clinical trials and personalized medicine.


Assuntos
Encéfalo/fisiopatologia , Modelos Animais de Doenças , Doença de Parkinson/fisiopatologia , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Doença de Parkinson/metabolismo
2.
Cell Tissue Res ; 373(1): 183-193, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29185072

RESUMO

The abnormal accumulation of α-synuclein aggregates in neurons, nerve fibers, or glial cells is the hallmark of a group of neurodegenerative diseases known collectively as α-synucleinopathies. Clinical, neuropathological, and experimental evidence strongly suggests that α-synuclein plays a role not only as a trigger of pathological processes at disease inception, but also as a mediator of pathological spreading during disease progression. Specific properties of α-synuclein, such as its ability to pass from one neuron to another, its tendency to aggregate, and its potential to generate self-propagating species, have been described and elucidated in animal models and may contribute to the relentless exacerbation of Parkinson's disease pathology in patients. Animal models used for studying α-synuclein accumulation, aggregation, and propagation are mostly based on three approaches: (1) intra-parenchymal inoculations of exogenous α-synuclein (e.g., synthetic α-synuclein fibrils), (2) transgenic mice, and (3) animals (mice or rats) in which α-synuclein overexpression is induced by viral vector injections. Whereas pathological α-synuclein changes are consistently observed in these models, important differences are also found. In particular, pronounced pathology in transgenic mice and viral vector-injected animals does not appear to involve self-propagating α-synuclein species. A critical discussion of these models reveals their strengths and limitations and provides the basis for recommendations concerning their use for future investigations.


Assuntos
alfa-Sinucleína/metabolismo , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Vetores Genéticos/metabolismo , Humanos
3.
Acta Neuropathol ; 133(3): 381-393, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28012041

RESUMO

Detection of α-synuclein lesions in peripheral tissues is a feature of human synucleinopathies of likely pathogenetic relevance and bearing important clinical implications. Experiments were carried out to elucidate the relationship between α-synuclein accumulation in the brain and in peripheral organs, and to identify potential pathways involved in long-distance protein transfer. Results of this in vivo study revealed a route-specific transmission of α-synuclein from the rat brain to the stomach. Following targeted midbrain overexpression of human α-synuclein, the exogenous protein was capable of reaching the gastric wall where it was accumulated into preganglionic vagal terminals. This brain-to-stomach connection likely involved intra- and inter-neuronal transfer of non-fibrillar α-synuclein that first reached the medulla oblongata, then gained access into cholinergic neurons of the dorsal motor nucleus of the vagus nerve and finally traveled via efferent fibers of these neurons contained within the vagus nerve. Data also showed a particular propensity of vagal motor neurons and efferents to accrue α-synuclein and deliver it to peripheral tissues; indeed, following its midbrain overexpression, human α-synuclein was detected within gastric nerve endings of visceromotor but not viscerosensory vagal projections. Thus, the dorsal motor nucleus of the vagus nerve represents a key relay center for central-to-peripheral α-synuclein transmission, and efferent vagal fibers may act as unique conduits for protein transfer. The presence of α-synuclein in peripheral tissues could reflect, at least in some synucleinopathy patients, an ongoing pathological process that originates within the brain and, from there, reaches distant organs innervated by motor vagal projections.


Assuntos
Fibras Autônomas Pré-Ganglionares/metabolismo , Encéfalo/metabolismo , Mucosa Gástrica/metabolismo , Nervo Vago/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/citologia , Colina O-Acetiltransferase/metabolismo , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neurônios/metabolismo , Gânglio Nodoso/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transdução Genética , Nervo Vago/fisiologia , alfa-Sinucleína/genética
4.
Brain ; 139(Pt 3): 856-70, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26719384

RESUMO

Aggregation and neuron-to-neuron transmission are attributes of α-synuclein relevant to its pathogenetic role in human synucleinopathies such as Parkinson's disease. Intraparenchymal injections of fibrillar α-synuclein trigger widespread propagation of amyloidogenic protein species via mechanisms that require expression of endogenous α-synuclein and, possibly, its structural corruption by misfolded conformers acting as pathological seeds. Here we describe another paradigm of long-distance brain diffusion of α-synuclein that involves inter-neuronal transfer of monomeric and/or oligomeric species and is independent of recruitment of the endogenous protein. Targeted expression of human α-synuclein was induced in the mouse medulla oblongata through an injection of viral vectors into the vagus nerve. Enhanced levels of intra-neuronal α-synuclein were sufficient to initiate its caudo-rostral diffusion that likely involved at least one synaptic transfer and progressively reached specific brain regions such as the locus coeruleus, dorsal raphae and amygdala in the pons, midbrain and forebrain. Transfer of human α-synuclein was compared in two separate lines of α-synuclein-deficient mice versus their respective wild-type controls and, interestingly, lack of endogenous α-synuclein expression did not counteract diffusion but actually resulted in a more pronounced and advanced propagation of exogenous α-synuclein. Self-interaction of adjacent molecules of human α-synuclein was detected in both wild-type and mutant mice. In the former, interaction of human α-synuclein with mouse α-synuclein was also observed and might have contributed to differences in protein transmission. In wild-type and α-synuclein-deficient mice, accumulation of human α-synuclein within recipient axons in the pons, midbrain and forebrain caused morphological evidence of neuritic pathology. Tissue sections from the medulla oblongata and pons were stained with different antibodies recognizing oligomeric, fibrillar and/or total (monomeric and aggregated) α-synuclein. Following viral vector transduction, monomeric, oligomeric and fibrillar protein was detected within donor neurons in the medulla oblongata. In contrast, recipient axons in the pons were devoid of immunoreactivity for fibrillar α-synuclein, indicating that non-fibrillar forms of α-synuclein were primarily transferred from one neuron to the other, diffused within the brain and led to initial neuronal injury. This study elucidates a paradigm of α-synuclein propagation that may play a particularly important role under pathophysiological conditions associated with enhanced α-synuclein expression. Rapid long-distance diffusion and accumulation of monomeric and oligomeric α-synuclein does not necessarily involve pathological seeding but could still result in a significant neuronal burden during the pathogenesis of neurodegenerative diseases.


Assuntos
Encéfalo/metabolismo , alfa-Sinucleína/biossíntese , Animais , Encéfalo/patologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , alfa-Sinucleína/deficiência , alfa-Sinucleína/genética
5.
Neurobiol Dis ; 78: 100-14, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25818009

RESUMO

Phosphorylation of the α-synuclein (α-syn) protein at Ser129 [P(S129)-α-syn] was found to be the most abundant form in intracellular inclusions in brains from Parkinson's disease (PD) patients. This finding suggests that P(S129)-α-syn plays a central role in the pathogenesis of PD. However, it is at present unclear whether P(S129)-α-syn is pathogenic driving the neurodegenerative process. Rodent studies using neither the phosphomimics of human α-syn nor co-expression of human wild-type α-syn and kinases phosphorylating α-syn at Ser129 gave consistent results. One major concern in interpreting these findings is that human α-syn was expressed above physiological levels inducing neurodegeneration in rat nigral neurons. In order to exclude this confounding factor, we took a different approach and increased the phosphorylation level of endogenous α-syn. For this purpose, we took advantage of recombinant adeno-associated viral (rAAV) vectors to deliver polo-like kinase (PLK) 2 or PLK3 in the substantia nigra and investigated whether increased levels of P(S129)-α-syn compromised the function and survival of nigral dopaminergic neurons. Interestingly, we observed that hyperphosphorylated α-syn did not induce nigral dopaminergic cell death, as assessed at 1 and 4months. Furthermore, histological analysis did not show any accumulation of α-syn protein or formation of inclusions. Using in vivo microdialysis, we found that the only measurable functional alteration was the depolarisation-induced release of dopamine, while the in vivo synthesis rate of DOPA and dopamine baseline release remained unaltered. Taken together, our results suggest that phosphorylation of α-syn at Ser129 does not confer a toxic gain of function per se.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Neurônios Dopaminérgicos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Sobrevivência Celular , Dependovirus , Dopamina/metabolismo , Feminino , Vetores Genéticos/efeitos adversos , Fosforilação , Ratos , Ratos Sprague-Dawley , Serina/metabolismo , Substância Negra/virologia
6.
Hum Mol Genet ; 21(14): 3173-92, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22513881

RESUMO

Although α-synuclein (α-SYN) aggregation is a hallmark of sporadic and familial Parkinson's disease (PD), it is not known how it contributes to early events of PD pathogenesis such as oxidative and inflammatory stress. Here, we addressed this question in a new animal model based on stereotaxic delivery of an adeno-associated viral vector (rAAV) for expression of human α-SYN in the ventral midbrain of mice lacking the transcription factor Nrf2 (Nrf2(-/-)). Two months after surgery, Nrf2(-/-) mice exhibited exacerbated degeneration of nigral dopaminergic neurons and increased dystrophic dendrites, reminiscent of Lewy neurites, which correlated with impaired proteasome gene expression and activity. Dopaminergic neuron loss was associated with an increase in neuroinflammation and gliosis that were intensified in Nrf2(-/-) mice. In response to exogenously added α-SYN, Nrf2(-/-) microglia failed to activate the expression of two anti-inflammatory genes, heme oxygenase-1 (HO-1) and nicotinamide adenine dinucleotide phosphate quinone oxidorreductase-1 (NQO1). This impaired Nrf2 response correlated with a shift in the microglial activation profile, towards increased production of proinflammatory markers, IL-6, IL-1ß and iNOS and reduced phagocytic capacity of fluorescent beads, and lower messenger RNA levels for TAM receptors Axl and Mer. Postmortem brain tissue samples from patients in early- to middle-stage progression of PD showed increased HO-1 expression in astrocytes and microglia, suggesting an attempt of the diseased brain to compensate these hallmarks of PD through activation of the Nrf2 pathway. This study demonstrates that α-SYN and Nrf2 deficiency cooperate on protein aggregation, neuroinflammation and neuronal death and provides a bifactorial animal model to study early-stage PD.


Assuntos
Fator 2 Relacionado a NF-E2/deficiência , Neurônios/citologia , Doença de Parkinson/genética , Doença de Parkinson/imunologia , alfa-Sinucleína/genética , Animais , Morte Celular , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica , Heme Oxigenase-1/genética , Heme Oxigenase-1/imunologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/imunologia , Neurônios/imunologia , Doença de Parkinson/patologia , Doença de Parkinson/fisiopatologia , alfa-Sinucleína/química , alfa-Sinucleína/imunologia
7.
Hum Mol Genet ; 21(4): 874-89, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22076440

RESUMO

Genetic studies have implicated the neuronal ubiquitin C-terminal hydrolase (UCH) protein UCH-L1 in Parkinson's disease (PD) pathogenesis. Moreover, the function of UCH-L1 may be lost in the brains of PD and Alzheimer's disease patients. We have previously reported that the UCH-L1 polymorphic variant S18Y, potentially protective against PD in population studies, demonstrates specific antioxidant functions in cell culture. Albeit genetic, biochemical and neuropathological data support an association between UCH-L1, PD, synaptic degeneration and oxidative stress, the relationship between the dopaminergic system and UCH-L1 status remains obscure. In the current study, we have examined the dopaminergic system of mice lacking endogenous UCH-L1 protein (gracile axonal dystrophy mice). Our findings show that the lack of wild-type (WT) UCH-L1 does not influence to any significant degree the dopaminergic system at baseline or following injections of the neurotoxin methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Furthermore, using a novel intrastriatal adenoviral injection protocol, we have found that mouse nigral neurons retrogradely transduced with S18Y UCH-L1, but not the WT protein, are significantly protected against MPTP toxicity. Overall, these data provide evidence for an antioxidant and neuroprotective effect of the S18Y variant of UCH-L1, but not of the WT protein, in the dopaminergic system, and may have implications for the pathogenesis of PD or related neurodegenerative conditions, in which oxidative stress might play a role.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Fármacos Neuroprotetores , Polimorfismo Genético/genética , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Adenoviridae/genética , Animais , Antioxidantes/metabolismo , Morte Celular/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Vetores Genéticos/genética , Humanos , Intoxicação por MPTP/patologia , Intoxicação por MPTP/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/citologia , Neostriado/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/metabolismo , Ubiquitina Tiolesterase/deficiência
8.
Proc Natl Acad Sci U S A ; 107(29): 13159-64, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20615977

RESUMO

Drug-induced dyskinesias in dopamine-denervated animals are known to depend on both pre- and postsynaptic changes of the nigrostriatal circuitry. In lesion models used thus far, changes occur in both of these compartments and, therefore, it has not been possible to dissect the individual contribution of each compartment in the pathophysiology of dyskinesias. Here we silenced the nigrostriatal dopamine neurotransmission without affecting the anatomical integrity of the presynaptic terminals using a short-hairpin RNA-mediated knockdown of tyrosine hydroxylase enzyme (shTH). This treatment resulted in significant reduction (by about 70%) in extracellular dopamine concentration in the striatum as measured by on-line microdialysis. Under these conditions, the animals remained nondyskinetic after chronic L-DOPA treatment, whereas partial intrastriatal 6-hydoxydopamine lesioned rats with comparable reduction in extracellular dopamine levels developed dyskinesias. On the other hand, apomorphine caused moderate to severe dyskinesias in both groups. Importantly, single-dose L-DOPA challenge in apomorphine-primed shTH animals failed to activate the already established abnormal postsynaptic responses. Taken together, these data provide direct evidence that the status of the presynaptic, DA releasing compartment is a critical determinant of both the induction and maintenance of L-DOPA-induced dyskinesias.


Assuntos
Dopamina/metabolismo , Discinesia Induzida por Medicamentos/metabolismo , Terminações Pré-Sinápticas/metabolismo , Animais , Apomorfina/farmacologia , Cromatografia Líquida de Alta Pressão , Dependovirus/genética , Suscetibilidade a Doenças , Regulação para Baixo/efeitos dos fármacos , Discinesia Induzida por Medicamentos/patologia , Feminino , Técnicas de Silenciamento de Genes , Levodopa , Microdiálise , Neostriado/efeitos dos fármacos , Neostriado/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Recombinação Genética/genética , Substância Negra/efeitos dos fármacos , Substância Negra/enzimologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Cells ; 12(4)2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36831238

RESUMO

Neuron-to-neuron transfer of pathogenic α-synuclein species is a mechanism of likely relevance to Parkinson's disease development. Experimentally, interneuronal α-synuclein spreading from the low brainstem toward higher brain regions can be reproduced by the administration of AAV vectors encoding for α-synuclein into the mouse vagus nerve. The aim of this study was to determine whether α-synuclein's spreading ability is shared by other proteins. Given α-synuclein synaptic localization, experiments involved intravagal injections of AAVs encoding for other synaptic proteins, ß-synuclein, VAMP2, or SNAP25. Administration of AAV-VAMP2 or AAV-SNAP25 caused robust transduction of either of the proteins in the dorsal medulla oblongata but was not followed by interneuronal VAMP2 or SNAP25 transfer and caudo-rostral spreading. In contrast, AAV-mediated ß-synuclein overexpression triggered its spreading to more frontal brain regions. The aggregate formation was investigated as a potential mechanism involved in protein spreading, and consistent with this hypothesis, results showed that overexpression of ß-synuclein, but not VAMP2 or SNAP25, in the dorsal medulla oblongata was associated with pronounced protein aggregation. Data indicate that interneuronal protein transfer is not a mere consequence of increased expression or synaptic localization. It is rather promoted by structural/functional characteristics of synuclein proteins that likely include their tendency to form aggregate species.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Camundongos , Animais , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Doença de Parkinson/metabolismo , Encéfalo/metabolismo , Tronco Encefálico/patologia , Proteína 2 Associada à Membrana da Vesícula/metabolismo
10.
Turk Gogus Kalp Damar Cerrahisi Derg ; 31(3): 352-357, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37664778

RESUMO

Background: The aim of this study was to investigate the prevalence of novel coronavirus disease 2019 (COVID-19) in patients hospitalized with primary spontaneous pneumothorax and to evaluate its possible effects on the clinical course, treatment, and the prognosis. Methods: Between April 2020 and January 2021, a total of 86 patients (78 males, 8 females; mean age: 27±5 years; range, 16 to 40 years) who had no underlying lung disease and were diagnosed with the first episode of spontaneous pneumothorax were retrospectively analyzed. At the same time of diagnosis, all patients were screened for COVID-19 via polymerase chain reaction test of nasopharyngeal swabs. According to the test results, the patients were divided into two groups as COVID-19(+) and COVID-19(-). The duration of air leak, hospital stay, recurrence rates and treatment modalities, and mortality rates of the two groups were compared. Results: Following a pneumothorax diagnosis, 18 (21%) patients were diagnosed with COVID-19. In COVID-19(+) patients, the mean air leak and lung expansion duration were significantly longer (p<0.0001 for both). In these patients, the mean length of hospital stay was also significantly longer (p<0.0001). During the median follow-up of six months, no mortality was observed and the recurrence rate was similar between the two groups (p=0.998). Conclusion: Our study results suggest that COVID-19 negatively affects the recovery time in patients with spontaneous pneumothorax.

11.
Neurobiol Dis ; 47(3): 367-77, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22659302

RESUMO

Impairments in the capacity of dopaminergic neurons to handle cytoplasmic dopamine may be a critical factor underlying the selective vulnerability of midbrain dopamine neurons in Parkinson's disease. Furthermore, toxicity of α-synuclein in dopaminergic neurons has been suggested to be mediated by direct interaction between dopamine and α-synuclein through formation of abnormal α-synuclein species, although direct in vivo evidence to support this hypothesis is lacking. Here, we investigated the role of dopamine availability on α-synuclein mediated neurodegeneration in vivo. We found that overexpression of α-synuclein in nigral dopamine neurons in mice with deficient vesicular storage of dopamine led to a significant increase in dopaminergic neurodegeneration. Importantly, silencing the tyrosine hydroxylase enzyme - thereby reducing dopamine content in the nigral neurons - reversed the increased vulnerability back to the baseline level observed in wild-type littermates, but failed to eliminate it completely. Importantly, TH knockdown was not effective in altering the toxicity in the wild-type animals. Taken together, our data suggest that under normal circumstances, in healthy dopamine neurons, cytoplasmic dopamine is tightly controlled such that it does not contribute significantly to α-synuclein mediated toxicity. Dysregulation of the dopamine machinery in the substantia nigra, on the other hand, could act as a trigger for induction of increased toxicity in these neurons and could explain how these neurons become more vulnerable and die in the disease process.


Assuntos
Dopamina/metabolismo , Neurônios/metabolismo , Substância Negra/citologia , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Análise de Variância , Animais , Cromatografia Líquida de Alta Pressão , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Eletroquímica , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Humanos , Ácido Hidroxi-Indolacético/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serotonina/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/genética
12.
Brain ; 134(Pt 8): 2302-11, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21712347

RESUMO

The neuroprotective effect of the glial cell line-derived neurotrophic factor has been extensively studied in various toxic models of Parkinson's disease. However, it remains unclear whether this neurotrophic factor can protect against the toxicity induced by the aggregation-prone protein α-synuclein. Targeted overexpression of human wild-type α-synuclein in the nigrostriatal system, using adeno-associated viral vectors, causes a progressive degeneration of the nigral dopamine neurons and the development of axonal pathology in the striatum. In the present study, we investigated, using different paradigms of delivery, whether glial cell line-derived neurotrophic factor can protect against the neurodegenerative changes and the cellular stress induced by α-synuclein. We found that viral vector-mediated delivery of glial cell line-derived neurotrophic factor into substantia nigra and/or striatum, administered 2-3 weeks before α-synuclein, was inefficient in preventing the wild-type α-synuclein-induced loss of dopamine neurons and terminals. In addition, glial cell line-derived neurotrophic factor overexpression did not ameliorate the behavioural deficit in this rat model of Parkinson's disease. Quantification of striatal α-synuclein-positive aggregates revealed that glial cell line-derived neurotrophic factor had no effect on α-synuclein aggregation. These data provide the evidence for the lack of neuroprotective effect of glial cell line-derived neurotrophic factor against the toxicity of human wild-type α-synuclein in an in vivo model of Parkinson's disease. The difference in neuroprotective efficacy of glial cell line-derived neurotrophic factor seen in our model and the commonly used neurotoxin models of Parkinson's disease, raises important issues pertinent to the interpretation of the results obtained in preclinical models of Parkinson's disease, and their relevance for the therapeutic use glial cell line-derived neurotrophic factor in patients with Parkinson's disease.


Assuntos
Fator Neurotrófico Derivado de Linhagem de Célula Glial/administração & dosagem , Proteínas de Filamentos Intermediários/metabolismo , Doenças Neurodegenerativas/prevenção & controle , Anfetamina/farmacologia , Animais , Animais Geneticamente Modificados , Contagem de Células , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Vetores Genéticos/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Proteínas de Fluorescência Verde/genética , Humanos , Proteínas de Filamentos Intermediários/genética , Doenças Neurodegenerativas/etiologia , Doença de Parkinson/complicações , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
13.
Ann Thorac Surg ; 113(5): e363-e365, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34331933

RESUMO

Surgical procedure is still used as the first choice in the treatment of pulmonary hydatid cysts. Video-assisted thoracoscopic surgery has started to be performed as a minimally invasive surgical option in recent years for lung hydatid cyst; however, few cases have been reported in the literature, especially in children. Thoracoscopic surgery experience is limited in pulmonary hydatid cyst for both adults and children. We present a pediatric case of giant lung hydatid cyst, in which we performed partial capitonnage with video-assisted thoracoscopic surgery and fibrin glue.


Assuntos
Equinococose Pulmonar , Adesivo Tecidual de Fibrina , Adulto , Criança , Equinococose Pulmonar/diagnóstico por imagem , Equinococose Pulmonar/cirurgia , Adesivo Tecidual de Fibrina/uso terapêutico , Humanos , Procedimentos Cirúrgicos Minimamente Invasivos , Estudos Retrospectivos , Cirurgia Torácica Vídeoassistida/métodos
14.
Sci Adv ; 8(35): eabn0356, 2022 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-36044566

RESUMO

Interneuronal transfer and brain spreading of pathogenic proteins are features of neurodegenerative diseases. Pathophysiological conditions and mechanisms affecting this spreading remain poorly understood. This study investigated the relationship between neuronal activity and interneuronal transfer of α-synuclein, a Parkinson-associated protein, and elucidated mechanisms underlying this relationship. In a mouse model of α-synuclein brain spreading, hyperactivity augmented and hypoactivity attenuated protein transfer. Important features of neuronal hyperactivity reported here were an exacerbation of oxidative and nitrative reactions, pronounced accumulation of nitrated α-synuclein, and increased protein aggregation. Data also pointed to mitochondria as key targets and likely sources of reactive oxygen and nitrogen species within hyperactive neurons. Rescue experiments designed to counteract the increased burden of reactive oxygen species reversed hyperactivity-induced α-synuclein nitration, aggregation, and interneuronal transfer, providing first evidence of a causal link between these pathological effects of neuronal stimulation and indicating a mechanistic role of oxidant stress in hyperactivity-induced α-synuclein spreading.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Encéfalo/metabolismo , Camundongos , Neurônios/metabolismo , Oxidantes , Doença de Parkinson/metabolismo
15.
Exp Mol Med ; 54(12): 2148-2161, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36473937

RESUMO

The clinical progression of neurodegenerative diseases correlates with the spread of proteinopathy in the brain. The current understanding of the mechanism of proteinopathy spread is far from complete. Here, we propose that inflammation is fundamental to proteinopathy spread. A sequence variant of α-synuclein (V40G) was much less capable of fibril formation than wild-type α-synuclein (WT-syn) and, when mixed with WT-syn, interfered with its fibrillation. However, when V40G was injected intracerebrally into mice, it induced aggregate spreading even more effectively than WT-syn. Aggregate spreading was preceded by sustained microgliosis and inflammatory responses, which were more robust with V40G than with WT-syn. Oral administration of an anti-inflammatory agent suppressed aggregate spreading, inflammation, and behavioral deficits in mice. Furthermore, exposure of cells to inflammatory cytokines increased the cell-to-cell propagation of α-synuclein. These results suggest that the inflammatory microenvironment is the major driver of the spread of synucleinopathy in the brain.


Assuntos
Doenças Neurodegenerativas , Sinucleinopatias , Camundongos , Animais , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Inflamação , Modelos Animais de Doenças
16.
J Mol Biol ; 433(12): 166961, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33774037

RESUMO

Neurotransmission relies on the tight spatial and temporal regulation of the synaptic vesicle (SV) cycle. Nerve terminals contain hundreds of SVs that form tight clusters. These clusters represent a distinct liquid phase in which one component of the phase are SVs and the other synapsin 1, a highly abundant synaptic protein. Another major family of disordered proteins at the presynapse includes synucleins, most notably α-synuclein. The precise physiological role of α-synuclein in synaptic physiology remains elusive, albeit its role has been implicated in nearly all steps of the SV cycle. To determine the effect of α-synuclein on the synapsin phase, we employ the reconstitution approach using natively purified SVs from rat brains and the heterologous cell system to generate synapsin condensates. We demonstrate that synapsin condensates recruit α-synuclein, and while enriched into these synapsin condensates, α-synuclein still maintains its high mobility. The presence of SVs enhances the rate of synapsin/α-synuclein condensation, suggesting that SVs act as catalyzers for the formation of synapsin condensates. Notably, at physiological salt and protein concentrations, α-synuclein alone is not able to cluster isolated SVs. Excess of α-synuclein disrupts the kinetics of synapsin/SV condensate formation, indicating that the molar ratio between synapsin and α-synuclein is important in assembling the functional condensates of SVs. Understanding the molecular mechanism of α-synuclein interactions at the nerve terminals is crucial for clarifying the pathogenesis of synucleinopathies, where α-synuclein, synaptic proteins and lipid organelles all accumulate as insoluble intracellular inclusions.


Assuntos
Encéfalo/citologia , Sinapsinas/metabolismo , Vesículas Sinápticas/metabolismo , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Células HEK293 , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Ratos , Sinapsinas/química , Transmissão Sináptica , alfa-Sinucleína/química , Proteína Vermelha Fluorescente
17.
Cell Death Differ ; 28(5): 1720-1732, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33323945

RESUMO

Alterations in the metabolism of iron and its accumulation in the substantia nigra pars compacta accompany the pathogenesis of Parkinson's disease (PD). Changes in iron homeostasis also occur during aging, which constitutes a PD major risk factor. As such, mitigation of iron overload via chelation strategies has been considered a plausible disease modifying approach. Iron chelation, however, is imperfect because of general undesired side effects and lack of specificity; more effective approaches would rely on targeting distinctive pathways responsible for iron overload in brain regions relevant to PD and, in particular, the substantia nigra. We have previously demonstrated that the Transferrin/Transferrin Receptor 2 (TfR2) iron import mechanism functions in nigral dopaminergic neurons, is perturbed in PD models and patients, and therefore constitutes a potential therapeutic target to halt iron accumulation. To validate this hypothesis, we generated mice with targeted deletion of TfR2 in dopaminergic neurons. In these animals, we modeled PD with multiple approaches, based either on neurotoxin exposure or alpha-synuclein proteotoxic mechanisms. We found that TfR2 deletion can provide neuroprotection against dopaminergic degeneration, and against PD- and aging-related iron overload. The effects, however, were significantly more pronounced in females rather than in males. Our data indicate that the TfR2 iron import pathway represents an amenable strategy to hamper PD progression. Data also suggest, however, that therapeutic strategies targeting TfR2 should consider a potential sexual dimorphism in neuroprotective response.


Assuntos
Fármacos Neuroprotetores/uso terapêutico , Doença de Parkinson/genética , Receptores da Transferrina/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Identidade de Gênero , Humanos , Camundongos , Fármacos Neuroprotetores/farmacologia
18.
Cell Rep ; 36(11): 109697, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525371

RESUMO

Midbrain dopaminergic (mDA) neurons are diverse in their projection targets, effect on behavior, and susceptibility to neurodegeneration. Little is known about the molecular mechanisms establishing this diversity during development. We show that the transcription factor BCL11A is expressed in a subset of mDA neurons in the developing and adult murine brain and in a subpopulation of pluripotent-stem-cell-derived human mDA neurons. By combining intersectional labeling and viral-mediated tracing, we demonstrate that Bcl11a-expressing mDA neurons form a highly specific subcircuit within the murine dopaminergic system. In the substantia nigra, the Bcl11a-expressing mDA subset is particularly vulnerable to neurodegeneration upon α-synuclein overexpression or oxidative stress. Inactivation of Bcl11a in murine mDA neurons increases this susceptibility further, alters the distribution of mDA neurons, and results in deficits in skilled motor behavior. In summary, BCL11A defines mDA subpopulations with highly distinctive characteristics and is required for establishing and maintaining their normal physiology.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Proteínas Repressoras/metabolismo , Animais , Comportamento Animal , Encéfalo/metabolismo , Dopamina/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Substância Negra/metabolismo , Substância Negra/patologia , Transcriptoma , Área Tegmentar Ventral/metabolismo , Área Tegmentar Ventral/patologia , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
19.
Eur J Neurosci ; 32(3): 409-22, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20704592

RESUMO

Lewy bodies, which are a pathological hallmark of Parkinson's disease, contain insoluble polymers of alpha-synuclein (alphasyn). Among the different modifications that can promote the formation of toxic alphasyn species, C-terminal truncation is among the most abundant alterations in patients with Parkinson's disease. In vitro, C-terminal truncated alphasyn aggregates faster and sub-stoichiometric amounts of C-terminal truncated alphasyn promote aggregation of the full-length alphasyn (alphasynFL) and induce neuronal toxicity. To address in vivo the putative stimulation of alphasyn-induced pathology by the presence of truncated alphasyn, we used recombinant adeno-associated virus to express either alphasynFL or a C-terminal truncated alphasyn (1-110) in rats. We adjusted the recombinant adeno-associated virus vector concentrations so that either protein alone led to only mild to moderate axonal pathology in the terminals of nigrostriatal dopamine neurons without frank cell loss. When these two forms of alphasyn were co-expressed at these pre-determined levels, it resulted in a more aggressive pathology in fiber terminals as well as dopaminergic cell loss in the substantia nigra. Using an antibody that did not detect the C-terminal truncated alphasyn (1-110) but only alphasynFL, we demonstrated that the co-expressed truncated protein promoted the progressive accumulation of alphasynFL and formation of larger pathological accumulations. Moreover, in the co-expression group, three of the eight animals showed apomorphine-induced turning, suggesting prominent post-synaptic alterations due to impairments in the dopamine release, whereas the mild pathology induced by either form alone did not cause motor abnormalities. Taken together these data suggest that C-terminal truncated alphasyn can interact with and exacerbate the formation of pathological accumulations containing alphasynFL in vivo.


Assuntos
Corpo Estriado/patologia , Neurônios/patologia , Substância Negra/patologia , alfa-Sinucleína/metabolismo , Análise de Variância , Animais , Corpo Estriado/metabolismo , Dependovirus/metabolismo , Dopamina/metabolismo , Imuno-Histoquímica , Corpos de Lewy/metabolismo , Corpos de Lewy/patologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
20.
Mol Ther ; 17(9): 1574-84, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19584816

RESUMO

Short-hairpin RNA (shRNA)-mediated gene knockdown is a powerful tool for targeted gene silencing and an emerging novel therapeutic strategy. Recent publications, however, reported unexpected toxicity after utilizing viral-mediated shRNA knockdown in vivo. Thus, it is currently unclear whether shRNA-mediated knockdown strategy can be used as a safe and efficient tool for gene silencing. In this study, we have generated rAAV vectors expressing shRNAs targeting the rat tyrosine hydroxylase (TH) mRNA (shTH) for testing the efficacy of in vivo TH knockdown in the nigral dopaminergic neurons. At high titers, not only the shTH vectors but also the scrambled and green fluorescence protein (GFP)-only controls caused cell death. In a dose-response study, we identified a dose window leading to >60% decrease in TH(+) neurons without any change in vesicular monoamine transporter-2 (VMAT2) expression. Moreover, using the safe and efficient dose, we showed that dopamine (DA) synthesis rate was significantly reduced and this lead to emergence of motor deficits in the shTH-expressing rats. Interestingly, these animals showed very robust and long-lasting recovery after a single systemic L-3,4-dihydroxyphenylalanine (L-DOPA) administration beyond what can be achieved in 6-hydroxydopamine (6-OHDA)-lesioned rats. Our results have implications for both mechanistic and therapeutic studies utilizing long-term shRNA-mediated gene silencing in the nigrostriatal projection system.


Assuntos
Adenoviridae/genética , Neurônios/metabolismo , Interferência de RNA/fisiologia , Substância Negra/citologia , Animais , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Dopamina/metabolismo , Feminino , Inativação Gênica/fisiologia , Vetores Genéticos/genética , Humanos , Levodopa , Ratos , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Tirosina 3-Mono-Oxigenase/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA