Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Gen Virol ; 105(3)2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38546100

RESUMO

Rift Valley fever virus (RVFV) is an emerging arboviral disease with pandemic potential. While infection is often self-limiting, a subset of individuals may develop late-onset encephalitis, accounting for up to 20 % of severe cases. Importantly, individuals displaying neurologic disease have up to a 53 % case fatality rate, yet the neuropathogenesis of RVFV infection remains understudied. In this study, we evaluated whether ex vivo postnatal rat brain slice cultures (BSCs) could be used to evaluate RVFV infection in the central nervous system. BSCs mounted an inflammatory response after slicing, which resolved over time, and they were viable in culture for at least 12 days. Infection of rat BSCs with pathogenic RVFV strain ZH501 induced tissue damage and apoptosis over 48 h. Viral replication in BSCs reached up to 1×107 p.f.u. equivalents/ml, depending on inoculation dose. Confocal immunofluorescent microscopy of cleared slices confirmed direct infection of neurons as well as activation of microglia and astrocytes. Further, RVFV-infected rat BSCs produced antiviral cytokines and chemokines, including MCP-1 and GRO/KC. This study demonstrates that rat BSCs support replication of RVFV for ex vivo studies of neuropathogenesis. This allows for continued and complementary investigation into RVFV infection in an ex vivo postnatal brain slice culture format.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Ratos , Animais , Vírus da Febre do Vale do Rift/fisiologia , Citocinas , Encéfalo , Morte Celular
2.
Acta Neuropathol ; 141(3): 359-381, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33496840

RESUMO

Accumulation of the tau protein in fibrillar intracellular aggregates is a defining feature of multiple neurodegenerative diseases collectively referred to as tauopathies. Despite intensive study of tau, there is limited information on the formation and clearance dynamics of tau inclusions. Using rAAV vectors to mediate expression of Dendra2-tagged human wild-type, P301L and pro-aggregant P301L/S320F tau proteins, with and without the addition of exogenous tau fibrillar seeds, we evaluated tau inclusion dynamics in organotypic brain slice culture (BSC) models using long-term optical pulse labeling methodology. Our studies reveal that tau inclusions typically form in 12-96 h in tauopathy BSC models. Unexpectedly, we demonstrate appreciable turnover of tau within inclusions with an average half-life of ~ 1 week when inclusions are newly formed. When BSCs with inclusions are aged in culture for extended periods, tau inclusions continue to turnover, but their half-lives increase to ~ 2 weeks and ~ 3 weeks after 1 and 2 months in culture, respectively. Individual tau inclusions can be long-lived structures that can persist for months in these BSC models and for even longer in the human brain. However, our data indicate that tau inclusions, are not 'tombstones', but dynamic structures with appreciable turnover. Understanding the cellular processes mediating this inclusion turnover may lead to new therapeutic strategies that could reverse pathological tau inclusion formation.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Neurônios/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Animais , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Camundongos , Neurônios/patologia , Técnicas de Cultura de Órgãos , Tauopatias/patologia
3.
Acta Neuropathol ; 140(6): 831-849, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33021680

RESUMO

Alpha-synuclein (αSyn) preformed fibrils (PFF) induce endogenous αSyn aggregation leading to reduced synaptic transmission. Neuronal activity modulates release of αSyn; however, whether neuronal activity regulates the spreading of αSyn pathology remains elusive. Here, we established a hippocampal slice culture system from wild-type (WT) mice and found that both Ca2+ influx and the uptake of αSyn PFF were higher in the CA3 than in the CA1 sub-region. Pharmacologically enhancing neuronal activity substantially increased αSyn pathology in αSyn PFF-treated hippocampal or midbrain slice cultures and accelerated dopaminergic neuron degeneration. Consistently, neuronal hyperactivity promoted PFF trafficking along axons/dendrites within microfluidic chambers. Unexpectedly, enhancing neuronal activity in LRRK2 G2019S mutant slice cultures further increased αSyn pathology, especially with more Lewy body (LB) forming than in WT slice cultures. Finally, following injection of αSyn PFF and chemogenetic modulators into the dorsal striatum of WT mice, both motor behavior and αSyn pathology were exacerbated likely by enhancing neuronal activity, since they were ameliorated by reducing neuronal activity. Thus, a greater understanding of the impact of neuronal activity on αSyn aggregation and spreading, as well as dopaminergic neuronal vulnerability, may provide new therapeutic strategies for patients with LB disease (LBD).


Assuntos
Encéfalo/patologia , Degeneração Neural/patologia , Neurônios/fisiologia , Sinucleinopatias/patologia , alfa-Sinucleína/metabolismo , Animais , Axônios/patologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Humanos , Doença por Corpos de Lewy/patologia , Camundongos , Sinucleinopatias/metabolismo
4.
Int J Mol Sci ; 20(8)2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30991705

RESUMO

Oxidative stress causes severe tissue injury of the central nervous system in ischemic brain damage (IBD), traumatic brain injury (TBI) and neurodegenerative disorders. In this study, we used hydrogen peroxide (H2O2) to induce oxidative stress in organotypic brain slice cultures (OBSCs), and investigated the protective effects of oxidative stress-tolerant (OST) stem cells harvested from human exfoliated deciduous teeth (SHED) which were co-cultivated with OBSCs. Using presto blue assay and immunostaining, we demonstrated that both normal SHED and OST-SHED could prevent H2O2-induced cell death, and increase the numbers of mature neuron and neuronal progenitors in the hippocampus of OBSCs. During co-cultivation, OST-SHED, but not normal SHED, exhibited neuronal cell morphology and expressed neuronal markers. Results from ELISA showed that both normal SHED and OST-SHED significantly decreased oxidative DNA damage in H2O2-treated OBSCs. SHED could also produce neurotrophic factor BDNF (brain derived neurotrophic factor) and promoted the production of IL-6 in OBSCs. Although OST-SHED had lower cell viability, the neuronal protection of OST-SHED was significantly superior to that of normal SHED. Our findings suggest that SHED, especially OST-SHED, could prevent oxidative stress induced brain damage. OST-SHED can be explored as a new therapeutic tool for IBD, TBI and neurodegenerative disorders.


Assuntos
Encéfalo/citologia , Técnicas de Cocultura , Neurônios/citologia , Neuroproteção , Estresse Oxidativo , Células-Tronco/citologia , Dente Decíduo/citologia , Animais , Morte Celular , Sobrevivência Celular , Células Cultivadas , Criança , Feminino , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos Endogâmicos ICR , Neurogênese
5.
Basic Clin Neurosci ; 15(2): 221-232, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39228449

RESUMO

Introduction: The pia-arachnoid is a critical component of cerebrospinal fluid removal. It covers and invaginates into the brain parenchyma, and physiologic failure results in hydrocephalus and cerebral edema. The purpose of this study was to characterize the role of arachnoid within brain parenchyma and determine if water flux and solute transport are affected by these intra-parenchymal cells. Methods: An immortalized arachnoid rat cell line was used to seed 300-µm organotypic rat brain slices of 4-week-old rats. Fluid and tracer transport analyses were conducted following a 7-10 day intraparenchymal growth period. The development of an arachnoid brain slice model was characterized using diffusion chamber experiments to calculate permeability, diffusion coefficient, and flux. Results: Labeled rat arachnoid cells readily penetrated organotypic cultures for up to 10 days. A significant reduction of dye and water flux across arachnoid-impregnated brain slices was observed after 3 hours in the diffusion chamber. Permeability decreased in whole brain slices containing arachnoid cells compared to slices without arachnoid cells. In comparison, a significant reduction of dextran across all slices occurred when molecular weights increased from 40 to 70 kDa. Conclusion: Tracer and small molecule studies show that arachnoid cells' presence significantly impacts water's movement through brain parenchyma. Size differential experiments indicate that the permeability of solute changed substantially between 40 and 70 kDa, an essential marker of blood-CSF barrier definition. We have developed an arachnoid organotypic model that reveals their ability to alter permeability and transport.

6.
Acta Neuropathol Commun ; 12(1): 83, 2024 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822428

RESUMO

Human brain experimental models recapitulating age- and disease-related characteristics are lacking. There is urgent need for human-specific tools that model the complex molecular and cellular interplay between different cell types to assess underlying disease mechanisms and test therapies. Here we present an adapted ex vivo organotypic slice culture method using human post-mortem brain tissue cultured at an air-liquid interface to also study brain white matter. We assessed whether these human post-mortem brain slices recapitulate the in vivo neuropathology and if they are suitable for pathophysiological, experimental and pre-clinical treatment development purposes, specifically regarding leukodystrophies. Human post-mortem brain tissue and cerebrospinal fluid were obtained from control, psychiatric and leukodystrophy donors. Slices were cultured up to six weeks, in culture medium with or without human cerebrospinal fluid. Human post-mortem organotypic brain slice cultures remained viable for at least six weeks ex vivo and maintained tissue structure and diversity of (neural) cell types. Supplementation with cerebrospinal fluid could improve slice recovery. Patient-derived organotypic slice cultures recapitulated and maintained known in vivo neuropathology. The cultures also showed physiologic multicellular responses to lysolecithin-induced demyelination ex vivo, indicating their suitability to study intrinsic repair mechanisms upon injury. The slice cultures were applicable for various experimental studies, as multi-electrode neuronal recordings. Finally, the cultures showed successful cell-type dependent transduction with gene therapy vectors. These human post-mortem organotypic brain slice cultures represent an adapted ex vivo model suitable for multifaceted studies of brain disease mechanisms, boosting translation from human ex vivo to in vivo. This model also allows for assessing potential treatment options, including gene therapy applications. Human post-mortem brain slice cultures are thus a valuable tool in preclinical research to study the pathomechanisms of a wide variety of brain diseases in living human tissue.


Assuntos
Encéfalo , Técnicas de Cultura de Órgãos , Humanos , Encéfalo/patologia , Encéfalo/metabolismo , Masculino , Feminino , Idoso , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurônios/patologia , Substância Branca/patologia , Substância Branca/metabolismo
7.
Cells ; 12(23)2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-38067189

RESUMO

Clinical and pre-clinical studies of neuropsychiatric (NP) disorders show altered astrocyte properties and synaptic networks. These are refined during early postnatal developmental (PND) stages. Thus, investigating early brain maturational trajectories is essential to understand NP disorders. However, animal experiments are highly time-/resource-consuming, thereby calling for alternative methodological approaches. The function of MEGF10 in astrocyte-mediated synapse elimination (pruning) is crucial to refine neuronal networks during development and adulthood. To investigate the impact of MEGF10 during PND in the rat prefrontal cortex (PFC) and its putative role in brain disorders, we established and validated an organotypic brain slice culture (OBSC) system. Using Western blot, we characterized the expression of MEGF10 and the synaptic markers synaptophysin and PSD95 in the cortex of developing pups. We then combined immunofluorescent-immunohistochemistry with Imaris-supported 3D analysis to compare age- and sex-dependent astrocyte-mediated pruning within the PFC in pups and OBSCs. We thereby validated this system to investigate age-dependent astrocyte-mediated changes in pruning during PND. However, further optimizations are required to use OBSCs for revealing sex-dependent differences. In conclusion, OBSCs offer a valid alternative to study physiological astrocyte-mediated synaptic remodeling during PND and might be exploited to investigate the pathomechanisms of brain disorders with aberrant synaptic development.


Assuntos
Astrócitos , Encefalopatias , Ratos , Animais , Hipocampo/metabolismo , Sinapses/metabolismo , Córtex Pré-Frontal/metabolismo , Encéfalo , Encefalopatias/metabolismo
8.
Front Oncol ; 13: 976945, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36793608

RESUMO

Organotypic cultures of murine brain slices are well-established tools in neuroscience research, including electrophysiology studies, modeling neurodegeneration, and cancer research. Here, we present an optimized ex vivo brain slice invasion assay that models glioblastoma multiforme (GBM) cell invasion into organotypic brain slices. Using this model, human GBM spheroids can be implanted with precision onto murine brain slices and cultured ex vivo to allow tumour cell invasion into the brain tissue. Traditional top-down confocal microscopy allows for imaging of GBM cell migration along the top of the brain slice, but there is limited resolution of tumour cell invasion into the slice. Our novel imaging and quantification technique involves embedding stained brain slices into an agar block, re-sectioning the slice in the Z-direction onto slides, and then using confocal microscopy to image cellular invasion into the brain tissue. This imaging technique allows for the visualization of invasive structures beneath the spheroid that would otherwise go undetected using traditional microscopy approaches. Our ImageJ macro (BraInZ) allows for the quantification of GBM brain slice invasion in the Z-direction. Importantly, we note striking differences in the modes of motility observed when GBM cells invade into Matrigel in vitro versus into brain tissue ex vivo highlighting the importance of incorporating the brain microenvironment when studying GBM invasion. In summary, our version of the ex vivo brain slice invasion assay improves upon previously published models by more clearly differentiating between migration along the top of the brain slice versus invasion into the slice.

9.
Methods Mol Biol ; 2623: 61-71, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36602679

RESUMO

During development of the cerebral cortex, neuroepithelial and radial glial cells undergo an oscillatory nuclear movement throughout their cell cycle, termed interkinetic nuclear migration. The nucleus of postmitotic neurons derived from these neural stem cells also translocates in a saltatory manner to enable neuronal migration toward the cortical plate. In these processes, various molecular motors, including cytoplasmic dynein, myosin II, and kinesins, are the driving force for nuclear migration at different stages. Despite efforts made to understand the mechanism regulating cortical development over decades, novel gene mutations discovered in neurodevelopmental disorders indicate that missing pieces still remain. Gene manipulation by in utero electroporation combined with live microscopy of neural stem cells in brain slices provides a powerful method to capture their detailed behaviors during proliferation and migration. The procedures described in this chapter enable the monitoring of cell cycle progression, mitosis, morphological changes, and migratory patterns in situ. This approach facilitates the elucidation of gene functions in cortical development and neurodevelopmental disorders.


Assuntos
Dineínas , Células-Tronco Neurais , Dineínas/genética , Dineínas/metabolismo , Microscopia , Córtex Cerebral , Neurônios/metabolismo , Movimento Celular/fisiologia , Eletroporação/métodos
10.
Yale J Biol Med ; 85(4): 501-21, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23239951

RESUMO

The brain is the most intricate, energetically active, and plastic organ in the body. These features extend to its cellular elements, the neurons and glia. Understanding neurons, or nerve cells, at the cellular and molecular levels is the cornerstone of modern neuroscience. The complexities of neuron structure and function require unusual methods of culture to determine how aberrations in or between cells give rise to brain dysfunction and disease. Here we review the methods that have emerged over the past century for culturing neurons in vitro, from the landmark finding by Harrison (1910) - that neurons can be cultured outside the body - to studies utilizing culture vessels, micro-islands, Campenot and brain slice chambers, and microfluidic technologies. We conclude with future prospects for neuronal culture and considerations for advancement. We anticipate that continued innovation in culture methods will enhance design capabilities for temporal control of media and reagents (chemotemporal control) within sub-cellular environments of three-dimensional fluidic spaces (microfluidic devices) and materials (e.g., hydrogels). They will enable new insights into the complexities of neuronal development and pathology.


Assuntos
Microfluídica , Neurônios/citologia , Células Cultivadas , Humanos
11.
Neurosci Lett ; 772: 136483, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35101576

RESUMO

This experiment mainly explored the protective effect and regulatory mechanism of melatonin (MEL) through its receptor on central nervous system (CNS) inflammation induced by lipopolysaccharide (LPS). The experiment was first divided into the following four groups: control group (CTRL group), LPS-induced inflammation model group (LPS group), LPS-treated MEL group (LPS + MEL group), and MEL administration group (MEL group). Later, a luzindole-antagonized LPS-MEL cotreatment group (LPS + MEL + LUZ group) was added to clarify the experimental results. ELISA was used to determine the inflammatory factor levels IL-6, IL-1ß, and IL-10 in brain slices. Western blotting was used to determine the expression levels of the microglia-specific protein CD11b and melatonin receptors MT1 and MT2 in brain slices. A large amount of IL-6 release and increased expression of CD11b protein were detected 24 h after inflammatory stimulation, while pretreatment with MEL inhibited the release of IL-6 and increased the expression of CD11b. At the same time, LPS induction downregulated the relative protein expression levels of MT1 and MT2. In addition, compared with the CTRL group and the LPS + MEL group, the administration of LUZ inhibited the protein expression of MT1. It increased the release of IL-1ß and IL-10, further indicating that MEL can alleviate LPS-induced neuroinflammation through the MT1 response. In short, MEL can reduce the neuroinflammatory response induced by LPS and exhibit related protective effects through MT1.


Assuntos
Anti-Inflamatórios/farmacologia , Encéfalo/efeitos dos fármacos , Melatonina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Encéfalo/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Interleucinas/metabolismo , Lipopolissacarídeos/toxicidade , Melatonina/metabolismo , Camundongos , Receptores de Melatonina/metabolismo
12.
Viruses ; 13(4)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916225

RESUMO

Measles virus (MV) can cause severe acute diseases as well as long-lasting clinical deteriorations due to viral-induced immunosuppression and neuronal manifestation. How the virus enters the brain and manages to persist in neuronal tissue is not fully understood. Various mutations in the viral genes were found in MV strains isolated from patient brains. In this study, reverse genetics was used to introduce mutations in the fusion, matrix and polymerase genes of MV. The generated virus clones were characterized in cell culture and used to infect rat brain slice cultures. A mutation in the carboxy-terminal domain of the matrix protein (R293Q) promoted the production of progeny virions. This effect was observed in Vero cells irrespective of the expression of the signaling lymphocyte activation molecule (SLAM). Furthermore, a mutation in the fusion protein (I225M) induced syncytia formation on Vero cells in the absence of SLAM and promoted viral spread throughout the rat brain slices. In this study, a solid ex vivo model was established to elucidate the MV mutations contributing to neural manifestation.


Assuntos
Encéfalo/virologia , Vírus do Sarampo/genética , Mutação , Neurônios/virologia , Proteínas Virais/genética , Tropismo Viral/genética , Animais , Chlorocebus aethiops , Células HEK293 , Humanos , Técnicas In Vitro , Sarampo/virologia , Vírus do Sarampo/patogenicidade , Vírus do Sarampo/fisiologia , Ratos Endogâmicos Lew , Genética Reversa , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Células Vero , Proteínas Virais de Fusão/genética
13.
Viruses ; 13(8)2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34452447

RESUMO

Measles virus (MV) and canine distemper virus (CDV) are closely related members of the family Paramyxoviridae, genus Morbillivirus. MV infection of humans and non-human primates (NHPs) results in a self-limiting disease, which rarely involves central nervous system (CNS) complications. In contrast, infection of carnivores with CDV usually results in severe disease, in which CNS complications are common and the case-fatality rate is high. To compare the neurovirulence and neurotropism of MV and CDV, we established a short-term organotypic brain slice culture system of the olfactory bulb, hippocampus, or cortex obtained from NHPs, dogs, and ferrets. Slices were inoculated ex vivo with wild-type-based recombinant CDV or MV expressing a fluorescent reporter protein. The infection level of both morbilliviruses was determined at different times post-infection. We observed equivalent infection levels and identified microglia as main target cells in CDV-inoculated carnivore and MV-inoculated NHP brain tissue slices. Neurons were also susceptible to MV infection in NHP brain slice cultures. Our findings suggest that MV and CDV have comparable neurotropism and intrinsic capacity to infect CNS-resident cells of their natural host species.


Assuntos
Encéfalo/virologia , Vírus da Cinomose Canina/fisiologia , Vírus do Sarampo/fisiologia , Tropismo Viral , Animais , Encéfalo/citologia , Cinomose/virologia , Vírus da Cinomose Canina/patogenicidade , Cães , Furões , Especificidade de Hospedeiro , Humanos , Sarampo/virologia , Microglia/virologia , Neurônios/virologia , Técnicas de Cultura de Órgãos , Primatas
14.
Noro Psikiyatr Ars ; 57(4): 333-337, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33354128

RESUMO

In vitro and in vivo models are efficiently used systems in neuroscience research to study the brain in normal or pathological conditions. There are many advantages to these systems, yet they also have significant limitations. In vitro cell cultures offer the opportunity to investigate the cell basics or primary response of a cell population against any treatment. However, these models do not always predict in vivo behavior. In vivo animal studies constitute the most realistic platform for research and therapeutic approaches, yet they are laborious, open to secondary complications and painful or stressful for the animals from an ethical point of view. Organotypic brain slice cultures provide an in vivo-like environment since they maintain three-dimensional cytoarchitecture of the brain thus enable to study many cell types in one system and allow precise control of the microenvironment. In this review, we will focus on the history and key features of organotypic brain slice cultures as well as its preparation.

15.
Front Cell Neurosci ; 14: 592005, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33473245

RESUMO

Microglia are central nervous system (CNS) resident immune cells that have been implicated in neuroinflammatory pathogenesis of a variety of neurological conditions. Their manifold context-dependent contributions to neuroinflammation are only beginning to be elucidated, which can be attributed in part to the challenges of studying microglia in vivo and the lack of tractable in vitro systems to study microglia function. Organotypic brain slice cultures offer a tissue-relevant context that enables the study of CNS resident cells and the analysis of brain slice microglial phenotypes has provided important insights, in particular into neuroprotective functions. Here we use RNA sequencing, direct digital quantification of gene expression with nCounter® technology and targeted analysis of individual microglial signature genes, to characterize brain slice microglia relative to acutely-isolated counterparts and 2-dimensional (2D) primary microglia cultures, a widely used in vitro surrogate. Analysis using single cell and population-based methods found brain slice microglia exhibited better preservation of canonical microglia markers and overall gene expression with stronger fidelity to acutely-isolated adult microglia, relative to in vitro cells. We characterized the dynamic phenotypic changes of brain slice microglia over time, after plating in culture. Mechanical damage associated with slice preparation prompted an initial period of inflammation, which resolved over time. Based on flow cytometry and gene expression profiling we identified the 2-week timepoint as optimal for investigation of microglia responses to exogenously-applied stimuli as exemplified by treatment-induced neuroinflammatory changes observed in microglia following LPS, TNF and GM-CSF addition to the culture medium. Altogether these findings indicate that brain slice cultures provide an experimental system superior to in vitro culture of microglia as a surrogate to investigate microglia functions, and the impact of soluble factors and cellular context on their physiology.

16.
Methods Mol Biol ; 2143: 133-144, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32524477

RESUMO

Organotypic hippocampal slice cultures (OHSCs) retain in vivo-like neuronal architecture, synaptic connections, and resident cell populations but gain in vitro advantages of accessibility to experimental manipulation and observation. This chapter describes how to prepare OHSCs from neonatal mice to study mechanisms of neuronal damage, including synapse loss and quantifying Aß-containing axonal swellings from Alzheimer's disease transgenic mice.


Assuntos
Encefalopatias/patologia , Hipocampo/fisiologia , Técnicas de Cultura de Órgãos/métodos , Animais , Animais Recém-Nascidos , Western Blotting , Dissecação/métodos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Microscopia Intravital/métodos , Proteínas Luminescentes/análise , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Microtomia/métodos , Proteínas do Tecido Nervoso/análise , Técnicas de Cultura de Órgãos/instrumentação
17.
J Parkinsons Dis ; 10(4): 1397-1410, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32716318

RESUMO

BACKGROUND: Synucleinopathy is a group of neurodegenerative disorders characterized by neurodegeneration and accumulation of alpha-synuclein (α-syn) aggregates in various brain regions. The detailed mechanism of α-syn-caused neurotoxicity remains obscure, which is partly due to the lack of a suitable model that retains the in vivo three-dimensional cellular network and allows a convenient dissection of the neurotoxic pathways. Recent studies revealed that the pre-formed recombinant α-syn amyloid fibrils (PFFs) induce a robust accumulation of pathogenic α-syn species in cultured cells and animals. OBJECTIVE: Our goal is to determine whether PFFs are able to induce the pathogenic α-syn accumulation and neurotoxicity in organotypic brain slice culture, an ex vivo system that retains the in vivo three-dimensional cell-cell connections. METHODS/RESULTS: Adding PFFs to cultured wild-type rat or mouse brain slices induced a time-dependent accumulation of pathogenic α-syn species, which was indicated by α-syn phosphorylated at serine 129 (pα-syn). The PFF-induced pα-syn was abolished in brain slices prepared from α-syn null mice, suggesting that the pα-syn is from the phosphorylation of endogenous α-syn. Human PFFs also induced pα-syn in brain slices prepared from mice expressing human α-syn on a mouse α-syn-null background. Furthermore, the synaptophysin immunoreactivity was inversely associated with pα-syn accumulation and an increase of neuronal loss was detected. CONCLUSION: PFF-treatment of brain slices is able to induce key pathological features of synucleinopathy: pα-syn accumulation and neurotoxicity. This model will be useful for investigating the neurotoxic mechanism and evaluating efficacy of therapeutic approaches.


Assuntos
Amiloide/metabolismo , Cerebelo/metabolismo , Lobo Frontal/metabolismo , Sinucleinopatias/metabolismo , alfa-Sinucleína/metabolismo , Animais , Cerebelo/patologia , Feminino , Lobo Frontal/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Fosforilação/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Sinucleinopatias/patologia
18.
Cancers (Basel) ; 12(6)2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560230

RESUMO

A challenge in cancer research is the definition of reproducible, reliable, and practical models, which reflect the effects of complex treatment modalities and the heterogeneous response of patients. Proton beam radiotherapy (PBRT), relative to conventional photon-based radiotherapy, offers the potential for iso-effective tumor control, while protecting the normal tissue surrounding the tumor. However, the effects of PBRT on the tumor microenvironment and the interplay with newly developed chemo- and immunotherapeutic approaches are still open for investigation. This work evaluated thin-cut tumor slice cultures (TSC) of head and neck cancer and organotypic brain slice cultures (OBSC) of adult mice brain, regarding their relevance for translational radiooncology research. TSC and OBSC were treated with PBRT and investigated for cell survival with a lactate dehydrogenase (LDH) assay, DNA repair via the DNA double strand break marker γH2AX, as well as histology with regards to morphology. Adult OBSC failed to be an appropriate model for radiobiological research questions. However, histological analysis of TSC showed DNA damage and tumor morphological results, comparable to known in vivo and in vitro data, making them a promising model to study novel treatment approaches in patient-derived xenografts or primary tumor material.

19.
Mol Brain ; 12(1): 45, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-31060588

RESUMO

Glioblastoma exhibits phenotypic and genetic heterogeneity, aggressive invasiveness, therapeutic resistance, and tumor recurrence, which can be explained by the existence of glioma stem cells (GSCs). In this study, we visualized the spatiotemporal dynamics of invasion of human GSCs in an orthotopic xenograft mouse model using time-lapse imaging of organotypic brain slice cultures and three-dimensional imaging of optically cleared whole brains. GSCs implanted in the striatum exhibited directional migration toward axon bundles, perivascular area, and the subventricular zone around the inferior horn of the lateral ventricle. GSCs migrated in a helical pattern around axon bundles in the striatum and invaded broadly in both the rostral and caudal directions. GSCs in the corpus callosum migrated more rapidly and unidirectionally toward the contralateral side with pseudopod extension. These characteristics of GSC invasion shared histological features observed in glioblastoma patients. Spatiotemporal visualization techniques can contribute to the elucidation of the mechanisms underlying GSC invasion that may lead to the development of effective therapy for glioblastoma.


Assuntos
Neoplasias Encefálicas/patologia , Glioma/patologia , Células-Tronco Neoplásicas/patologia , Animais , Encéfalo/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Imageamento Tridimensional , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Imagem com Lapso de Tempo
20.
J Mol Neurosci ; 68(1): 29-37, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30806968

RESUMO

Acyl ghrelin, a novel brain-gut peptide, is an endogenous ligand for the growth hormone secretagogue receptor. Accumulated research data have shown that acyl ghrelin exercises a significant neuroprotective effect against cerebral ischemia/reperfusion (I/R) injury in animal models and in cultured neurons during the acute phase, usually, 1 day after cerebral ischemia. The chronic effects of acyl ghrelin 1 week after brain ischemia remain largely unknown. In this study, we explored the effects of acyl ghrelin on cultured organotypic brain slices and cortical neurons which were injured by oxygen-glucose deprivation/reperfusion(OGD/R) for 7 days. The underlying molecular mechanisms were deciphered based on label-free proteomic analysis. Acyl ghrelin treatment promoted neurite (axons and dendrites) growth and alleviated the neuronal damage in both cultured brain slices and neurons. Proteomic analysis showed that cell division control protein 42 (Cdc42) mediates the effects of acyl ghrelin on neurite growth. Acyl ghrelin stimulated the expression of Cdc42 and neurite growth in cultured neurons comparing with OGD/R group. Inhibition of Cdc42 attenuated the effects of acyl ghrelin. These results suggest that acyl ghrelin promotes neurite growth during the later stage after OGD/R injury via Cdc42. Our study suggests that acyl ghrelin may be promising to restore the neuronal structure in the late phase after stroke.


Assuntos
Isquemia Encefálica/metabolismo , Grelina/metabolismo , Crescimento Neuronal , Animais , Hipóxia Celular , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Grelina/farmacologia , Glucose/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa