Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Toxicol Environ Health A ; 87(1): 22-32, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-37818790

RESUMO

Perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) are polyfluoroalkyl substances (PFAS) used as surface coatings in manufacturing. Exposure to PFAS was shown to be correlated with infertility, low birth weight, and delayed aspects of pubertal development in mammals. Despite many correlational studies, there have been few direct investigations examining the link between PFAS exposure and early animal development. The aim of this study was to (1) examine the effects of PFOA on development and reproduction using the roundworm Caenorhabditis elegans, a model with a high predictive value for human reproductive toxicity and (2) compare observations to exposure to PFOS. PFAS exposure did not markedly alter egg hatching but delayed population growth, in part due to slower larval development. PFAS-exposed worms took longer to progress through larval stages to reach reproductive maturity, and this was not attributed to PFOA-induced toxicity to their food. Our results provide a robust benchmark for testing developmental and reproductive toxicity for other PFAS and PFAS-alternatives which continue to be used in manufacturing and released into the environment.


Assuntos
Ácidos Alcanossulfônicos , Fluorocarbonos , Animais , Humanos , Caenorhabditis elegans , Crescimento Demográfico , Fluorocarbonos/toxicidade , Ácidos Alcanossulfônicos/toxicidade , Mamíferos
2.
Front Cell Neurosci ; 17: 1271169, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38089145

RESUMO

AMPA receptors (AMPARs) mediate the majority of fast excitatory transmission in the brain. Regulation of AMPAR levels at synapses controls synaptic strength and underlies information storage and processing. Many proteins interact with the intracellular domain of AMPARs to regulate their trafficking and synaptic clustering. However, a growing number of extracellular factors important for glutamatergic synapse development, maturation and function have emerged that can also regulate synaptic AMPAR levels. This mini-review highlights extracellular protein factors that regulate AMPAR trafficking to control synapse development and plasticity. Some of these factors regulate AMPAR clustering and mobility by interacting with the extracellular N-terminal domain of AMPARs whereas others regulate AMPAR trafficking indirectly via their respective signaling receptors. While several of these factors are secreted from neurons, others are released from non-neuronal cells such as glia and muscle. Although it is apparent that secreted factors can act locally on neurons near their sites of release to coordinate individual synapses, it is less clear if they can diffuse over longer ranges to coordinate related synapses within a circuit or region of the brain. Given that there are hundreds of factors that can be secreted from neuronal and non-neuronal cells, it will not be surprising if more extracellular factors that modulate AMPARs and glutamatergic synapses are discovered. Many open questions remain including where and when the factors are expressed, what regulates their secretion from different cell types, what controls their diffusion, stability, and range of action, and how their cognate receptors influence intracellular signaling to control AMPAR trafficking.

3.
MicroPubl Biol ; 20232023.
Artigo em Inglês | MEDLINE | ID: mdl-37179968

RESUMO

We identified the Low-Density Lipoprotein (LDL) Receptor Related Protein-2 (LRP-2) in a RNAi screen for genes that regulate glutamatergic behavior in C. elegans . lrp-2 loss-of-function mutants have defects in glutamatergic mechanosensory nose-touch behavior and suppress increased spontaneous reversals induced by GLR-1(A/T), a constitutively-active form of the AMPA-type glutamate receptor GLR-1. Total and surface levels of GLR-1 are increased throughout the ventral nerve cord of lrp-2 mutants suggesting that LRP-2 promotes glutamatergic signaling by regulating some aspect of GLR-1 trafficking, localization or function.

4.
J Undergrad Neurosci Educ ; 22(1): A1-A13, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38322408

RESUMO

Compared to traditional teaching laboratory activities, course-based undergraduate research experiences (CUREs) can increase student engagement and confidence, improve scientific literacy, enhance critical thinking, and promote accessibility in STEM. Here we describe a versatile CURE for an upper-level Neurobiology course that incorporates genetic, molecular, cellular, and behavioral experiments into a semester-long investigation to identify genes important for glutamate synapse formation or function in C. elegans. Following introduction to the CURE approach and basic C. elegans techniques, students construct their own low-cost optogenetics rigs, which we describe in detail here, to activate a mechanosensory escape reflex via photostimulation. They then perform a small-scale RNAi screen with this light-activated behavioral readout. Once a gene of interest is identified, students submit a proposal to investigate the role of this gene in nervous system function and spend the rest of the semester carrying out follow-up experiments using mutant strains. We also describe ways in which this CURE can be modified depending on the pedagogical objectives, availability of materials, or research interests of the instructor. Participating in this lab significantly enhanced students' abilities to see themselves as STEM professionals and prompted students to report substantial gains in skills critical for entry into and success in graduate and medical schools. In addition to the benefits CUREs provide to students, faculty benefit from the generation of preliminary data and training of students for potential independent research projects.

5.
MicroPubl Biol ; 20212021 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-34142022

RESUMO

Mechanosensory or chemosensory activation of glutamatergicASH amphid sensory neurons promotes avoidancebehaviors in C. elegans. Wormswith mutations in the transcription factor DMD-10 have impaired ASH-mediated sensorimotor reflexes. We hypothesized that the behavioral dysfunction in dmd-10 mutants could arise from impaired ASH development or survival leading to disrupted glutamatergic signaling.To test this, we performed in vivo fluorescence microscopy of young adult C. elegans amphid neurons after labeling with the lipophilic dye DiI. We quantified the number of ASH neurons as well as five other amphid sensory neuron pairs. We found that the number of amphid neurons in dmd-10 mutants was the same as in wild-type worms. Our results suggest that dmd-10 is not required for amphid neuron development or survival in mature C. elegans.

6.
PeerJ ; 9: e10892, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33665029

RESUMO

The Doublesex/Mab-3 Domain transcription factor DMD-10 is expressed in several cell types in C. elegans, including in the nervous system. We sought to investigate whether DMD-10 is required for normal neuronal function using behavioral assays. We found that mutation of dmd-10 did not broadly affect behavior. dmd-10 mutants were normal in several behavioral assays including a body bends assay for locomotion, egg laying, chemotaxis and response to gentle touch to the body. dmd-10 mutants did have defects in nose-touch responsiveness, which requires the glutamate receptor GLR-1. However, using quantitative fluorescence microscopy to measure levels of a GLR-1::GFP fusion protein in the ventral nerve cord, we found no evidence supporting a difference in the number of GLR-1 synapses or in the amount of GLR-1 present in dmd-10 mutants. dmd-10 mutants did have decreased responsiveness to high osmolarity, which, along with nose-touch, is sensed by the polymodal sensory neuron ASH. Furthermore, mutation of dmd-10 impaired behavioral response to optogenetic activation of ASH, suggesting that dmd-10 promotes neuronal signaling in ASH downstream of sensory receptor activation. Together our results suggest that DMD-10 is important in regulating the frequency of multiple ASH-dependent behavioral responses.

7.
PLoS One ; 16(2): e0245587, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33529210

RESUMO

Regulation of AMPA-type glutamate receptor (AMPAR) expression and function alters synaptic strength and is a major mechanism underlying synaptic plasticity. Although transcription is required for some forms of synaptic plasticity, the transcription factors that regulate AMPA receptor expression and signaling are incompletely understood. Here, we identify the Snail family transcription factor ces-1 in an RNAi screen for conserved transcription factors that regulate glutamatergic behavior in C. elegans. ces-1 was originally discovered as a selective cell death regulator of neuro-secretory motor neuron (NSM) and I2 interneuron sister cells in C. elegans, and has almost exclusively been studied in the NSM cell lineage. We found that ces-1 loss-of-function mutants have defects in two glutamatergic behaviors dependent on the C. elegans AMPA receptor GLR-1, the mechanosensory nose-touch response and spontaneous locomotion reversals. In contrast, ces-1 gain-of-function mutants exhibit increased spontaneous reversals, and these are dependent on glr-1 consistent with these genes acting in the same pathway. ces-1 mutants have wild type cholinergic neuromuscular junction function, suggesting that they do not have a general defect in synaptic transmission or muscle function. The effect of ces-1 mutation on glutamatergic behaviors is not due to ectopic cell death of ASH sensory neurons or GLR-1-expressing neurons that mediate one or both of these behaviors, nor due to an indirect effect on NSM sister cell deaths. Rescue experiments suggest that ces-1 may act, in part, in GLR-1-expressing neurons to regulate glutamatergic behaviors. Interestingly, ces-1 mutants suppress the increased reversal frequencies stimulated by a constitutively-active form of GLR-1. However, expression of glr-1 mRNA or GFP-tagged GLR-1 was not decreased in ces-1 mutants suggesting that ces-1 likely promotes GLR-1 function. This study identifies a novel role for ces-1 in regulating glutamatergic behavior that appears to be independent of its canonical role in regulating cell death in the NSM cell lineage.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ácido Glutâmico/metabolismo , Receptores de AMPA/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Proteínas de Caenorhabditis elegans/genética , Proteínas de Ligação a DNA/genética , Mutação com Ganho de Função , Interneurônios/metabolismo , Locomoção/genética , Mutação com Perda de Função , Plasticidade Neuronal/genética , Interferência de RNA , Células Receptoras Sensoriais/metabolismo , Fatores de Transcrição da Família Snail/genética , Sinapses/metabolismo , Fatores de Transcrição/genética , Transgenes
8.
PLoS Genet ; 17(2): e1009375, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33561120

RESUMO

Several intracellular trafficking pathways contribute to the regulation of AMPA receptor (AMPAR) levels at synapses and the control of synaptic strength. While much has been learned about these intracellular trafficking pathways, a major challenge is to understand how extracellular factors, such as growth factors, neuropeptides and hormones, impinge on specific AMPAR trafficking pathways to alter synaptic function and behavior. Here, we identify the secreted ligand PVF-1 and its cognate VEGF receptor homologs, VER-1 and VER-4, as regulators of glutamate signaling in C. elegans. Loss of function mutations in ver-1, ver-4, or pvf-1, result in decreased cell surface levels of the AMPAR GLR-1 and defects in glutamatergic behavior. Rescue experiments indicate that PVF-1 is expressed and released from muscle, whereas the VERs function in GLR-1-expressing neurons to regulate surface levels of GLR-1 and glutamatergic behavior. Additionally, ver-4 is unable to rescue glutamatergic behavior in the absence of pvf-1, suggesting that VER function requires endogenous PVF-1. Inducible expression of a pvf-1 rescuing transgene suggests that PVF-1 can function in the mature nervous system to regulate GLR-1 signaling. Genetic double mutant analysis suggests that the VERs act together with the VPS-35/retromer recycling complex to promote cell surface levels of GLR-1. Our data support a genetic model whereby PVF-1/VER signaling acts with retromer to promote recycling and cell surface levels of GLR-1 to control behavior.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Ácido Glutâmico/metabolismo , Fator de Crescimento Derivado de Plaquetas/genética , Receptores de AMPA/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Transdução de Sinais/genética , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Membrana Celular/metabolismo , Regulação da Expressão Gênica , Microscopia de Fluorescência , Mutação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de AMPA/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Células Receptoras Sensoriais/metabolismo , Sinapses/metabolismo
9.
J Neurosci ; 41(14): 3082-3093, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33622778

RESUMO

Reversible modification of AMPA receptors (AMPARs) with ubiquitin regulates receptor levels at synapses and controls synaptic strength. The conserved deubiquitinating enzyme (DUB) ubiquitin-specific protease-46 (USP-46) removes ubiquitin from AMPARs and protects them from degradation in both Caenorhabditis elegans and mammals. Although DUBs are critical for diverse physiological processes, the mechanisms that regulate DUBs, especially in the nervous system, are not well understood. We and others previously showed that the WD40-repeat proteins WDR-48 and WDR-20 bind to and stimulate the catalytic activity of USP-46. Here, we identify an activity-dependent mechanism that regulates WDR-20 expression and show that WDR-20 works together with USP-46 and WDR-48 to promote surface levels of the C. elegans AMPAR GLR-1. usp-46, wdr-48, and wdr-20 loss-of-function mutants exhibit reduced levels of GLR-1 at the neuronal surface and corresponding defects in GLR-1-mediated behavior. Increased expression of WDR-20, but not WDR-48, is sufficient to increase GLR-1 surface levels in an usp-46-dependent manner. Loss of usp-46, wdr-48, and wdr-20 function reduces the rate of local GLR-1 insertion in neurites, whereas overexpression of wdr-20 is sufficient to increase the rate of GLR-1 insertion. Genetic manipulations that chronically reduce or increase glutamate signaling result in reciprocal alterations in wdr-20 transcription and homeostatic compensatory changes in surface GLR-1 levels that are dependent on wdr-20 This study identifies wdr-20 as a novel activity-regulated gene that couples chronic changes in synaptic activity with increased local insertion and surface levels of GLR-1 via the DUB USP-46.SIGNIFICANCE STATEMENT Deubiquitinating enzymes (DUBs) are critical regulators of synapse development and function; however, the regulatory mechanisms that control their various physiological functions are not well understood. This study identifies a novel role for the DUB ubiquitin-specific protease-46 (USP-46) and its associated regulatory protein WD40-repeat protein-20 (WDR-20) in regulating local insertion of glutamate receptors into the neuronal cell surface. This work also identifies WDR-20 as an activity-regulated gene that couples chronic changes in synaptic activity with homeostatic compensatory increases in surface levels of GLR-1 via USP-46. Given that 35% of USP family DUBs associate with WDR proteins, understanding the mechanisms by which WDR proteins regulate USP-46 could have implications for a large number of DUBs in other cell types.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Enzimas Desubiquitinantes/metabolismo , Endopeptidases/metabolismo , Receptores de Glutamato/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte/genética , Membrana Celular/genética , Enzimas Desubiquitinantes/genética , Endopeptidases/genética , Receptores de Glutamato/genética
10.
Methods Mol Biol ; 1948: 183-198, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30771178

RESUMO

Accumulation of misfolded αSyn and mitochondrial dysfunction are central features of Parkinson's disease. Growing evidence points to a relationship between these two phenomena as oligomeric α-synuclein (αSyn) can interact with mitochondria and impair their function. Standardization of methods to prepare αSyn oligomers and isolate functional mitochondria will facilitate efforts to expand upon early findings. Here we present detailed protocols for preparing soluble αSyn oligomers; for isolating functional mitochondria from mouse tissue; and for simultaneously measuring several aspects of mitochondrial physiology. These protocols will benefit future studies aimed at characterizing the mitotoxicity of αSyn species isolated from the brains of synucleinopathy patients as well as efforts to identify small molecules and genetic or environmental alterations that prevent αSyn-induced mitochondrial dysfunction.


Assuntos
Mitocôndrias/metabolismo , Multimerização Proteica , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Animais , Encéfalo/metabolismo , Cálcio/metabolismo , Fígado/metabolismo , Camundongos , Doença de Parkinson/metabolismo , Dobramento de Proteína , Solubilidade
12.
Nat Commun ; 6: 7314, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26076669

RESUMO

ß-Sheet-rich α-synuclein (αS) aggregates characterize Parkinson's disease (PD). αS was long believed to be a natively unfolded monomer, but recent work suggests it also occurs in α-helix-rich tetramers. Crosslinking traps principally tetrameric αS in intact normal neurons, but not after cell lysis, suggesting a dynamic equilibrium. Here we show that freshly biopsied normal human brain contains abundant αS tetramers. The PD-causing mutation A53T decreases tetramers in mouse brain. Neurons derived from an A53T patient have decreased tetramers. Neurons expressing E46K do also, and adding 1-2 E46K-like mutations into the canonical αS repeat motifs (KTKEGV) further reduces tetramers, decreases αS solubility and induces neurotoxicity and round inclusions. The other three fPD missense mutations likewise decrease tetramer:monomer ratios. The destabilization of physiological tetramers by PD-causing missense mutations and the neurotoxicity and inclusions induced by markedly decreasing tetramers suggest that decreased α-helical tetramers and increased unfolded monomers initiate pathogenesis. Tetramer-stabilizing compounds should prevent this.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Doença de Parkinson/genética , alfa-Sinucleína/genética , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Immunoblotting , Imuno-Histoquímica , Células-Tronco Pluripotentes Induzidas , Camundongos , Mutação de Sentido Incorreto , Doença de Parkinson/metabolismo , Estrutura Quaternária de Proteína/genética , Ratos , Ratos Sprague-Dawley , alfa-Sinucleína/metabolismo
13.
Mol Biol Cell ; 26(10): 1887-900, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25788288

RESUMO

Regulation of glutamate receptor (GluR) abundance at synapses by clathrin-mediated endocytosis can control synaptic strength and plasticity. We take advantage of viable, null mutations in subunits of the clathrin adaptor protein 2 (AP2) complex in Caenorhabditis elegans to characterize the in vivo role of AP2 in GluR trafficking. In contrast to our predictions for an endocytic adaptor, we found that levels of the GluR GLR-1 are decreased at synapses in the ventral nerve cord (VNC) of animals with mutations in the AP2 subunits APM-2/µ2, APA-2/α, or APS-2/σ2. Rescue experiments indicate that APM-2/µ2 functions in glr-1-expressing interneurons and the mature nervous system to promote GLR-1 levels in the VNC. Genetic analyses suggest that APM-2/µ2 acts upstream of GLR-1 endocytosis in the VNC. Consistent with this, GLR-1 accumulates in cell bodies of apm-2 mutants. However, GLR-1 does not appear to accumulate at the plasma membrane of the cell body as expected, but instead accumulates in intracellular compartments including Syntaxin-13- and RAB-14-labeled endosomes. This study reveals a novel role for the AP2 clathrin adaptor in promoting the abundance of GluRs at synapses in vivo, and implicates AP2 in the regulation of GluR trafficking at an early step in the secretory pathway.


Assuntos
Complexo 2 de Proteínas Adaptadoras/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Sistema Nervoso/metabolismo , Receptores de AMPA/metabolismo , Sinapses/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Regulação da Expressão Gênica , Mutação , Transporte Proteico , Receptores de AMPA/genética , Sinapses/genética
14.
Biochemistry ; 54(2): 279-92, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25490121

RESUMO

Despite two decades of research, the structure-function relationships of endogenous, physiological forms of α-synuclein (αSyn) are not well understood. Most in vitro studies of this Parkinson's disease-related protein have focused on recombinant αSyn that is unfolded and monomeric, assuming that this represents its state in the normal human brain. Recently, we have provided evidence that αSyn exists in considerable part in neurons, erythrocytes, and other cells as a metastable multimer that principally sizes as a tetramer. In contrast to recombinant αSyn, physiological tetramers purified from human erythrocytes have substantial α-helical content and resist pathological aggregation into ß-sheet rich fibers. Here, we report the first method to fully purify soluble αSyn from the most relevant source, human brain. We describe protocols that purify αSyn to homogeneity from nondiseased human cortex using ammonium sulfate precipitation, gel filtration, and ion exchange, hydrophobic interaction, and affinity chromatographies. Cross-linking of the starting material and the partially purified chromatographic fractions revealed abundant αSyn multimers, including apparent tetramers, but these were destabilized in large part to monomers during the final purification step. The method also fully purified the homologue ß-synuclein, with a similar outcome. Circular dichroism spectroscopy showed that purified, brain-derived αSyn can display more helical content than the recombinant protein, but this result varied. Collectively, our data suggest that purifying αSyn to homogeneity destabilizes native, α-helix-rich multimers that exist in intact and partially purified brain samples. This finding suggests existence of a stabilizing cofactor (e.g., a small lipid) present inside neurons that is lost during final purification.


Assuntos
Química Encefálica , Estabilidade Proteica , alfa-Sinucleína/isolamento & purificação , Precipitação Química , Cromatografia em Gel , Cromatografia por Troca Iônica , Dicroísmo Circular , Humanos , Espectrometria de Massas , Multimerização Proteica , Estrutura Secundária de Proteína , alfa-Sinucleína/química , beta-Sinucleína/isolamento & purificação
15.
PLoS One ; 9(7): e103727, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25075858

RESUMO

A switch in the conformational properties of α-synuclein (αS) is hypothesized to be a key step in the pathogenic mechanism of Parkinson's disease (PD). Whereas the beta-sheet-rich state of αS has long been associated with its pathological aggregation in PD, a partially alpha-helical state was found to be related to physiological lipid binding; this suggests a potential role of the alpha-helical state in controlling synaptic vesicle cycling and resistance to ß-sheet rich aggregation. N-terminal acetylation is the predominant post-translational modification of mammalian αS. Using circular dichroism, isothermal titration calorimetry, and fluorescence spectroscopy, we have analyzed the effects of N-terminal acetylation on the propensity of recombinant human αS to form the two conformational states in interaction with lipid membranes. Small unilamellar vesicles of negatively charged lipids served as model membranes. Consistent with previous NMR studies using phosphatidylserine, we found that membrane-induced α-helical folding was enhanced by N-terminal acetylation and that greater exothermic heat could be measured upon vesicle binding of the modified protein. Interestingly, the folding and lipid binding enhancements with phosphatidylserine in vitro were weak when compared to that of αS with GM1, a lipid enriched in presynaptic membranes. The resultant increase in helical folding propensity of N-acetylated αS enhanced its resistance to aggregation. Our findings demonstrate the significance of the extreme N-terminus for folding nucleation, for relative GM1 specificity of αS-membrane interaction, and for a protective function of N-terminal-acetylation against αS aggregation mediated by GM1.


Assuntos
Gangliosídeo G(M1)/química , Processamento de Proteína Pós-Traducional , alfa-Sinucleína/química , Acetilação , Amiloide/química , Humanos , Agregados Proteicos , Ligação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Termodinâmica
16.
J Biol Chem ; 289(31): 21490-507, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24942732

RESUMO

α-Synuclein (αSyn) aggregation and mitochondrial dysfunction both contribute to the pathogenesis of Parkinson disease (PD). Although recent studies have suggested that mitochondrial association of αSyn may disrupt mitochondrial function, it is unclear what aggregation state of αSyn is most damaging to mitochondria and what conditions promote or inhibit the effect of toxic αSyn species. Because the neuronal populations most vulnerable in PD are characterized by large cytosolic Ca(2+) oscillations that burden mitochondria, we examined mitochondrial Ca(2+) stress in an in vitro system comprising isolated mitochondria and purified recombinant human αSyn in various aggregation states. Using fluorimetry to simultaneously measure four mitochondrial parameters, we observed that soluble, prefibrillar αSyn oligomers, but not monomeric or fibrillar αSyn, decreased the retention time of exogenously added Ca(2+), promoted Ca(2+)-induced mitochondrial swelling and depolarization, and accelerated cytochrome c release. Inhibition of the permeability transition pore rescued these αSyn-induced changes in mitochondrial parameters. Interestingly, the mitotoxic effects of αSyn were specifically dependent upon both electron flow through complex I and mitochondrial uptake of exogenous Ca(2+). Our results suggest that soluble prefibrillar αSyn oligomers recapitulate several mitochondrial phenotypes previously observed in animal and cell models of PD: complex I dysfunction, altered membrane potential, disrupted Ca(2+) homeostasis, and enhanced cytochrome c release. These data reveal how the association of oligomeric αSyn with mitochondria can be detrimental to the function of cells with high Ca(2+)-handling requirements.


Assuntos
Biopolímeros/fisiologia , Cálcio/fisiologia , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/fisiologia , alfa-Sinucleína/fisiologia , Animais , Benzotiazóis , Biopolímeros/química , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Tiazóis/metabolismo , alfa-Sinucleína/química
17.
J Biol Chem ; 288(9): 6371-85, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23319586

RESUMO

Aggregation of α-synuclein (αSyn) in neurons produces the hallmark cytopathology of Parkinson disease and related synucleinopathies. Since its discovery, αSyn has been thought to exist normally in cells as an unfolded monomer. We recently reported that αSyn can instead exist in cells as a helically folded tetramer that resists aggregation and binds lipid vesicles more avidly than unfolded recombinant monomers (Bartels, T., Choi, J. G., and Selkoe, D. J. (2011) Nature 477, 107-110). However, a subsequent study again concluded that cellular αSyn is an unfolded monomer (Fauvet, B., Mbefo, M. K., Fares, M. B., Desobry, C., Michael, S., Ardah, M. T., Tsika, E., Coune, P., Prudent, M., Lion, N., Eliezer, D., Moore, D. J., Schneider, B., Aebischer, P., El-Agnaf, O. M., Masliah, E., and Lashuel, H. A. (2012) J. Biol. Chem. 287, 15345-15364). Here we describe a simple in vivo cross-linking method that reveals a major ~60-kDa form of endogenous αSyn (monomer, 14.5 kDa) in intact cells and smaller amounts of ~80- and ~100-kDa forms with the same isoelectric point as the 60-kDa species. Controls indicate that the apparent 60-kDa tetramer exists normally and does not arise from pathological aggregation. The pattern of a major 60-kDa and minor 80- and 100-kDa species plus variable amounts of free monomers occurs endogenously in primary neurons and erythroid cells as well as neuroblastoma cells overexpressing αSyn. A similar pattern occurs for the homologue, ß-synuclein, which does not undergo pathogenic aggregation. Cell lysis destabilizes the apparent 60-kDa tetramer, leaving mostly free monomers and some 80-kDa oligomer. However, lysis at high protein concentrations allows partial recovery of the 60-kDa tetramer. Together with our prior findings, these data suggest that endogenous αSyn exists principally as a 60-kDa tetramer in living cells but is lysis-sensitive, making the study of natural αSyn challenging outside of intact cells.


Assuntos
Células Eritroides/metabolismo , Neurônios/metabolismo , Multimerização Proteica/fisiologia , alfa-Sinucleína/metabolismo , beta-Sinucleína/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , alfa-Sinucleína/genética , beta-Sinucleína/genética
18.
J Histochem Cytochem ; 60(1): 31-44, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22205679

RESUMO

Skeletal muscle growth and its regeneration following injury rely on myogenic progenitor cells, a heterogeneous population that includes the satellite cells and other interstitial progenitors. The present study demonstrates that surface expression of ß4 integrin marks a population of vessel-associated interstitial muscle progenitor cells. Muscle ß4 integrin-positive cells do not express myogenic markers upon isolation. However, they are capable of undergoing myogenic specification in vitro and in vivo: ß4 integrin cells differentiate into multinucleated myotubes in culture dishes and contribute to muscle regeneration upon delivery into diseased mice. Subfractionation of ß4 integrin-expressing cells based on CD31 expression does not further enrich for myogenic precursors. These findings support the expression of ß4 integrin in interstitial, vessel-associated cells with myogenic activity within adult skeletal muscle.


Assuntos
Integrina beta4/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Células-Tronco/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/transplante , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/inervação , Músculo Esquelético/patologia , Regeneração , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Células-Tronco/citologia
19.
J Neurosci ; 30(31): 10431-40, 2010 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-20685985

RESUMO

Although clinically distinct, schizophrenia and Alzheimer's disease are common and devastating disorders that profoundly impair cognitive function. For Alzheimer's disease, key mechanistic insights have emerged from genetic studies that identified causative mutations in amyloid precursor protein (APP) and presenilin. Several genes have been associated with schizophrenia and other major psychoses, and understanding their normal functions will help elucidate the underlying causes of these disorders. One such gene is disrupted-in-schizophrenia 1 (DISC1). DISC1 and APP have been implicated separately in cortical development, with each having roles in both neuronal migration and neurite outgrowth. Here, we report a previously unrecognized biochemical and functional interaction between DISC1 and APP. Using in utero electroporation in the living rat brain, we show that DISC1 acts downstream of APP and Disabled-1 to regulate cortical precursor cell migration. Specifically, overexpression of DISC1 rescues the migration defect caused by a loss of APP expression. Moreover, knockdown of APP in cultured embryonic neurons results in altered subcellular localization of DISC1. Using transfected cells and normal brain tissue, we show that APP and DISC1 coimmunoprecipitate and that the intracellular domain of APP interacts with the N-terminal domain of DISC1. Based on these findings, we hypothesize that the APP cytoplasmic region transiently interacts with DISC1 to help regulate the translocation of DISC1 to the centrosome, where it plays a key role in controlling neuronal migration during cortical development.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Western Blotting , Células Cultivadas , Córtex Cerebral/metabolismo , Eletroporação , Imunofluorescência , Imunoprecipitação , Microscopia Confocal , Proteínas do Tecido Nervoso/genética , Ratos , Ratos Sprague-Dawley , Transfecção
20.
J Cell Sci ; 121(Pt 9): 1426-34, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18397996

RESUMO

Although the contribution of bone marrow-derived cells to regenerating skeletal muscle has been repeatedly documented, there remains considerable debate as to whether this incorporation is exclusively a result of inflammatory cell fusion to regenerating myofibers or whether certain populations of bone marrow-derived cells have the capacity to differentiate into muscle. The present study uses a dual-marker approach in which GFP(+) cells were intravenously transplanted into lethally irradiated beta-galactosidase(+) recipients to allow for simple determination of donor and host contribution to the muscle. FACS analysis of cardiotoxin-damaged muscle revealed that CD45(+) bone-marrow side-population (SP) cells, a group enriched in hematopoietic stem cells, can give rise to CD45(-)/Sca-1(+)/desmin(+) cells capable of myogenic differentiation. Moreover, after immunohistochemical examination of the muscles of both SP- and whole bone marrow-transplanted animals, we noted the presence of myofibers composed only of bone marrow-derived cells. Our findings suggest that a subpopulation of bone marrow SP cells contains precursor cells whose progeny have the potential to differentiate towards a muscle lineage and are capable of de novo myogenesis following transplantation and initiation of muscle repair via chemical damage.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Desenvolvimento Muscular , Células-Tronco/citologia , Animais , Ataxina-1 , Ataxinas , Biomarcadores/metabolismo , Transplante de Medula Óssea , Contagem de Células , Linhagem da Célula , Separação Celular , Feminino , Proteínas de Fluorescência Verde/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Regeneração , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA