Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biomedicines ; 12(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38255199

RESUMO

Synapse loss is the principal cause of cognitive decline in Alzheimer's disease (AD) and related disorders (ADRD). Synapse development depends on the intricate dynamics of the neuronal cytoskeleton. Cofilin, the major protein regulating actin dynamics, can be sequestered into cofilactin rods, intra-neurite bundles of cofilin-saturated actin filaments that can disrupt vesicular trafficking and cause synaptic loss. Rods are a brain pathology in human AD and mouse models of AD and ADRD. Eliminating rods is the focus of this paper. One pathway for rod formation is triggered in ~20% of rodent hippocampal neurons by disease-related factors (e.g., soluble oligomers of Amyloid-ß (Aß)) and requires cellular prion protein (PrPC), active NADPH oxidase (NOX), and cytokine/chemokine receptors (CCRs). FDA-approved antagonists of CXCR4 and CCR5 inhibit Aß-induced rods in both rodent and human neurons with effective concentrations for 50% rod reduction (EC50) of 1-10 nM. Remarkably, two D-amino acid receptor-active peptides (RAP-103 and RAP-310) inhibit Aß-induced rods with an EC50 of ~1 pM in mouse neurons and ~0.1 pM in human neurons. These peptides are analogs of D-Ala-Peptide T-Amide (DAPTA) and share a pentapeptide sequence (TTNYT) antagonistic to several CCR-dependent responses. RAP-103 does not inhibit neuritogenesis or outgrowth even at 1 µM, >106-fold above its EC50. N-terminal methylation, or D-Thr to D-Ser substitution, decreases the rod-inhibiting potency of RAP-103 by 103-fold, suggesting high target specificity. Neither RAP peptide inhibits neuronal rod formation induced by excitotoxic glutamate, but both inhibit rods induced in human neurons by several PrPC/NOX pathway activators (Aß, HIV-gp120 protein, and IL-6). Significantly, RAP-103 completely protects against Aß-induced loss of mature and developing synapses and, at 0.1 nM, reverses rods in both rodent and human neurons (T½ ~ 3 h) even in the continuous presence of Aß. Thus, this orally available, brain-permeable peptide should be highly effective in reducing rod pathology in multifactorial neurological diseases with mixed proteinopathies acting through PrPC/NOX.

2.
Biomedicines ; 11(11)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-38001943

RESUMO

Cofilactin rod pathology, which can initiate synapse loss, has been extensively studied in rodent neurons, hippocampal slices, and in vivo mouse models of human neurodegenerative diseases such as Alzheimer's disease (AD). In these systems, rod formation induced by disease-associated factors, such as soluble oligomers of Amyloid-ß (Aß) in AD, utilizes a pathway requiring cellular prion protein (PrPC), NADPH oxidase (NOX), and cytokine/chemokine receptors (CCR5 and/or CXCR4). However, rod pathways have not been systematically assessed in a human neuronal model. Here, we characterize glutamatergic neurons differentiated from human-induced pluripotent stem cells (iPSCs) for the formation of rods in response to activators of the PrPC-dependent pathway. Optimization of substratum, cell density, and use of glial-conditioned medium yielded a robust system for studying the development of Aß-induced rods in the absence of glia, suggesting a cell-autonomous pathway. Rod induction in younger neurons requires ectopic expression of PrPC, but this dependency disappears by Day 55. The quantification of proteins within the rod-inducing pathway suggests that increased PrPC and CXCR4 expression may be factors in the doubling of the rod response to Aß between Days 35 and 55. FDA-approved antagonists to CXCR4 and CCR5 inhibit the rod response. Rods were predominantly observed in dendrites, although severe cytoskeletal disruptions prevented the assignment of over 40% of the rods to either an axon or dendrite. In the absence of glia, a condition in which rods are more readily observed, neurons mature and fire action potentials but do not form functional synapses. However, PSD95-containing dendritic spines associate with axonal regions of pre-synaptic vesicles containing the glutamate transporter, VGLUT1. Thus, our results identified stem cell-derived neurons as a robust model for studying cofilactin rod formation in a human cellular environment and for developing effective therapeutic strategies for the treatment of dementias arising from multiple proteinopathies with different rod initiators.

3.
J Neurosci Res ; 90(1): 229-42, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21932365

RESUMO

Inflammation accompanied by severe oxidative stress plays a vital role in the orchestration and progression of neurodegeneration prevalent in chronic and acute central nervous system pathologies as well as in aging. The proinflammatory cytokine tumor necrosis factor-α (TNFα) elicits the formation of the bioactive ceramide by stimulating the hydrolysis of the membrane lipid sphingomyelin by sphingomyelinase activities. Ceramide stimulates the formation of reactive oxygen species (ROS) and apoptotic mechanisms in both neurons and nonneuronal cells, establishing a link between sphingolipid metabolism and oxidative stress. We demonstrated in SH-SY5Y human neuroblastoma cells and primary cortical neurons that TNFα is a potent stimulator of Mg(2+) -dependent neutral sphingomyelinase (Mg(2+) -nSMase) activity, and sphingomyelin hydrolysis, rather than de novo synthesis, was the predominant source of ceramide increases. Mg(2+) -nSMase activity preceded an accumulation of ROS by a neuronal NADPH oxidase (NOX). Notably, TNFα provoked an NOX-dependent oxidative damage to sphingosine kinase-1, which generates sphingosine-1-phosphate, a ceramide metabolite associated with neurite outgrowth. Indeed, ceramide and ROS inhibited neurite outgrowth of dorsal root ganglion neurons by disrupting growth cone motility. Blunting ceramide and ROS formation both rescued sphingosine kinase-1 activity and neurite outgrowth. Our studies suggest that TNFα-mediated activation of Mg(2+) -nSMase and NOX in neuronal cells not only produced the neurotoxic intermediates ceramide and ROS but also directly antagonized neuronal survival mechanisms, thus accelerating neurodegeneration.


Assuntos
NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Fator de Necrose Tumoral alfa/toxicidade , Animais , Apoptose/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Ceramidas , Córtex Cerebral/citologia , Embrião de Galinha , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/fisiologia , Humanos , Magnésio/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neuroblastoma/patologia , Neurônios/citologia , Palmitatos/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio
4.
Cells ; 10(10)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34685706

RESUMO

Proteins of the actin depolymerizing factor (ADF)/cofilin family are ubiquitous among eukaryotes and are essential regulators of actin dynamics and function. Mammalian neurons express cofilin-1 as the major isoform, but ADF and cofilin-2 are also expressed. All isoforms bind preferentially and cooperatively along ADP-subunits in F-actin, affecting the filament helical rotation, and when either alone or when enhanced by other proteins, promotes filament severing and subunit turnover. Although self-regulating cofilin-mediated actin dynamics can drive motility without post-translational regulation, cells utilize many mechanisms to locally control cofilin, including cooperation/competition with other proteins. Newly identified post-translational modifications function with or are independent from the well-established phosphorylation of serine 3 and provide unexplored avenues for isoform specific regulation. Cofilin modulates actin transport and function in the nucleus as well as actin organization associated with mitochondrial fission and mitophagy. Under neuronal stress conditions, cofilin-saturated F-actin fragments can undergo oxidative cross-linking and bundle together to form cofilin-actin rods. Rods form in abundance within neurons around brain ischemic lesions and can be rapidly induced in neurites of most hippocampal and cortical neurons through energy depletion or glutamate-induced excitotoxicity. In ~20% of rodent hippocampal neurons, rods form more slowly in a receptor-mediated process triggered by factors intimately connected to disease-related dementias, e.g., amyloid-ß in Alzheimer's disease. This rod-inducing pathway requires a cellular prion protein, NADPH oxidase, and G-protein coupled receptors, e.g., CXCR4 and CCR5. Here, we will review many aspects of cofilin regulation and its contribution to synaptic loss and pathology of neurodegenerative diseases.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Fatores de Despolimerização de Actina/química , Sequência de Aminoácidos , Animais , Humanos , Neuritos/metabolismo , Neurogênese
5.
PLoS One ; 16(3): e0248309, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33705493

RESUMO

Nearly 50% of individuals with long-term HIV infection are affected by the onset of progressive HIV-associated neurocognitive disorders (HAND). HIV infiltrates the central nervous system (CNS) early during primary infection where it establishes persistent infection in microglia (resident macrophages) and astrocytes that in turn release inflammatory cytokines, small neurotoxic mediators, and viral proteins. While the molecular mechanisms underlying pathology in HAND remain poorly understood, synaptodendritic damage has emerged as a hallmark of HIV infection of the CNS. Here, we report that the HIV viral envelope glycoprotein gp120 induces the formation of aberrant, rod-shaped cofilin-actin inclusions (rods) in cultured mouse hippocampal neurons via a signaling pathway common to other neurodegenerative stimuli including oligomeric, soluble amyloid-ß and proinflammatory cytokines. Previous studies showed that synaptic function is impaired preferentially in the distal proximity of rods within dendrites. Our studies demonstrate gp120 binding to either chemokine co-receptor CCR5 or CXCR4 is capable of inducing rod formation, and signaling through this pathway requires active NADPH oxidase presumably through the formation of superoxide (O2-) and the expression of cellular prion protein (PrPC). These findings link gp120-mediated oxidative stress to the generation of rods, which may underlie early synaptic dysfunction observed in HAND.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Hipocampo/metabolismo , NADPH Oxidases/metabolismo , Neurônios/metabolismo , Proteínas PrPC/metabolismo , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Fatores de Despolimerização de Actina/genética , Actinas/genética , Animais , Proteína gp120 do Envelope de HIV/genética , Infecções por HIV/genética , HIV-1/genética , Camundongos , Camundongos Knockout , NADPH Oxidases/genética , Estresse Oxidativo/genética , Proteínas PrPC/genética , Receptores CCR5/genética , Receptores CXCR4/genética
6.
Mol Cell Neurosci ; 41(2): 274-85, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19344766

RESUMO

The proinflammatory cytokines TNFalpha and Il-1beta orchestrate the progression of CNS inflammation, which substantially contributes to neurodegeneration in many CNS pathologies. TNFalpha and Il-1beta stimulate actin filament reorganization in non-neuronal cells often accompanied by the formation of reactive oxygen species (ROS). Actin filament dynamics is vital for cellular plasticity, mitochondrial function, and gene expression despite being highly susceptible to oxidative damage. We demonstrated that, in neuronal cells, TNFalpha and Il-1beta stimulate a transient, redox-dependent reorganization of the actin cytoskeleton into lamellipodia under the regulation of Rac1 and a neuronal NADPH oxidase as the source of ROS. The persistent presence of intracellular ROS provoked oxidative damage (carbonylation) to actin coinciding with the loss of lamellipodia and arrest of cellular plasticity. Inhibition of NADPH oxidase activity or Rac1 abolished the adverse effects of cytokines. These findings suggest that oxidative damage to the neuronal actin cytoskeleton could represent a key step in CNS neurodegeneration.


Assuntos
Actinas/metabolismo , Citocinas , NADPH Oxidases/metabolismo , Neurônios , Estresse Oxidativo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Citocinas/farmacologia , Citoesqueleto/metabolismo , Humanos , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oxirredução , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
Adv Exp Med Biol ; 644: 232-49, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19209826

RESUMO

Dynamics of actin filaments is pivotal to many fundamental cellular processes such as Dcytokinesis, motility, morphology, vesicle and organelle transport, gene transcription and senescence. In vivo kinetics of actin filament dynamics is far from the equilibrium in vitro and these profound differences are attributed to large number of regulatory proteins. In particular, proteins of the ADF/cofilin family greatly increase actin filament dynamics by severing filaments and enhancing depolymerization of ADP-actin monomers from their pointed ends. Cofilin binds cooperatively to a minor conformer of F-actin in which the subunits are slightly under rotated along the filament helical axis. At high stoichiometry of cofilin to actin subunits, cofilin actually stabilizes actin filaments. Many isoforms oftropomyosin appear to compete with ADF/cofilin proteins for binding to actin filaments. Tropomyosin isoforms studied to date prefer binding to the "untwisted" conformer of F-actin and through their protection and stabilization of F-actin, recruit myosin II and assemble different actin superstructures from the cofilin-actin filaments. However, some tropomyosin isoforms may synergize with ADF/cofilin to enhance filament dynamics, suggesting that the different isoforms of tropomyosins, many of which show developmental or tissue specific expression profiles, play major roles in the assembly and turnover of actin superstructures. Different actin superstructures can overlap both spatially and temporally within a cell, but can be differentiated from each other based upon their kinetic and kinematic properties. Furthermore, local regulation of ADF/cofilin activity through signal transduction pathways could be one mechanism to alter the dynamic balance in F-actin-binding of certain tropomyosin isoforms in subcellular domains.


Assuntos
Fatores de Despolimerização de Actina/química , Cofilina 1/química , Tropomiosina/fisiologia , Fatores de Despolimerização de Actina/metabolismo , Actinas/química , Trifosfato de Adenosina/química , Animais , Ligação Competitiva , Citoesqueleto/metabolismo , Humanos , Proteínas dos Microfilamentos/química , Modelos Biológicos , Miosina Tipo II/química , Ligação Proteica , Isoformas de Proteínas , Tropomiosina/química
8.
Curr HIV Res ; 16(4): 258-269, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30280668

RESUMO

The implementation of combination antiretroviral therapy (cART) as the primary means of treatment for HIV infection has achieved a dramatic decline in deaths attributed to AIDS and the reduced incidence of severe forms of HIV-associated neurocognitive disorders (HAND) in infected individuals. Despite these advances, milder forms of HAND persist and prevalence of these forms of neurocognitive impairment are rising with the aging population of HIV infected individuals. HIV enters the CNS early in the pathophysiology establishing persistent infection in resident macrophages and glial cells. These infected cells, in turn, secrete neurotoxic viral proteins, inflammatory cytokines, and small metabolites thought to contribute to neurodegenerative processes. The viral envelope protein gp120 has been identified as a potent neurotoxin affecting neurodegeneration via indirect and direct mechanisms involving interactions with chemokine co-receptors CCR5 and CXCR4. This short review focuses on gp120 neurotropism and associated mechanisms of neurotoxicity linked to chemokine receptors CCR5 and CXCR4 with a new perspective on plasma membrane lipid rafts as an active participant in gp120-mediated neurodegeneration underlying HIV induced CNS pathology.


Assuntos
Nefropatia Associada a AIDS/fisiopatologia , Proteína gp120 do Envelope de HIV/toxicidade , Microdomínios da Membrana/metabolismo , Neurônios/patologia , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Humanos
9.
J Immunol Methods ; 462: 42-47, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30099015

RESUMO

Immunoglobulins (Ig) are proteins that preserve immune homeostasis and are quantified to infer changes to the acquired humoral immune response in mammals. Measuring Ig in non-model wildlife for immune surveillance often requires ingenuity, and rigorous standardization of methodologies to provide reliable results especially when lacking species-specific reagents. We modified and optimized existing ELISA methodology utilizing the binding properties of Staphylococcus-derived Protein A (PrtA) to immunoglobulin G (IgG). We enhanced the assay for quantifying IgG in Steller sea lion (SSL) serum using critical quality control measures including dilution linearity, spike and percent recoveries, and internal controls. Of the modifications made, heat treatment of SSL serum enhanced accuracy and precision of IgG measurements by improving linearity and percent recovery in parallel dilutions and serum spikes. Purified canine IgG standard was not affected by heat inactivation. These results support that confounding serum proteins interfere with binding of PrtA with IgG demonstrating the need for heat treatment of serum to optimize IgG quantification using the PrtA-ELISA. Further, essential validation measures ensure proper assay performance. Consequently, the improved PrtA-ELISA provides species-independent IgG detection with validation criteria to enhance accuracy and precision for addressing future immunological questions in non-model wildlife in clinical, ecological, and conservation contexts.


Assuntos
Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Animais , Cães , Ensaio de Imunoadsorção Enzimática/métodos , Leões-Marinhos
10.
Biomed Res Int ; 2014: 191767, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25050325

RESUMO

A persistent inflammatory and oxidative stress is a hallmark of most chronic CNS pathologies (Alzheimer's (ALS)) as well as the aging CNS orchestrated by the proinflammatory cytokines tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1ß). Loss of the integrity and plasticity of neuronal morphology and connectivity comprises an early step in neuronal degeneration and ultimate decline of cognitive function. We examined in vitro whether TNFα or IL-1ß impaired morphology and motility of growth cones in spinal cord neuron cultures. TNFα and IL-1ß paralyzed growth cone motility and induced growth cone collapse in a dose-dependent manner reflected by complete attenuation of neurite outgrowth. Scavenging reactive oxygen species (ROS) or inhibiting NADPH oxidase activity rescued loss of neuronal motility and morphology. TNFα and IL-1ß provoked rapid, NOX-mediated generation of ROS in advancing growth cones, which preceded paralysis of motility and collapse of morphology. Increases in ROS intermediates were accompanied by an aberrant, nonproductive reorganization of actin filaments. These findings suggest that NADPH oxidase serves as a pivotal source of oxidative stress in neurons and together with disruption of actin filament reorganization contributes to the progressive degeneration of neuronal morphology in the diseased or aging CNS.


Assuntos
Cones de Crescimento/patologia , Mediadores da Inflamação/toxicidade , Interleucina-1beta/toxicidade , Paralisia/patologia , Espécies Reativas de Oxigênio/toxicidade , Medula Espinal/patologia , Fator de Necrose Tumoral alfa/toxicidade , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Animais , Corpo Celular/efeitos dos fármacos , Corpo Celular/metabolismo , Embrião de Galinha , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/enzimologia , Humanos , Modelos Biológicos , NADPH Oxidases/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Oxirredução/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo
11.
Prion ; 8(6): 375-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25426519

RESUMO

Increasing evidence suggests that proteins exhibiting "prion-like" behavior cause distinct neurodegenerative diseases, including inherited, sporadic and acquired types. The conversion of cellular prion protein (PrP(C)) to its infectious protease resistant counterpart (PrP(Res)) is the essential feature of prion diseases. However, PrP(C) also performs important functions in transmembrane signaling, especially in neurodegenerative processes. Beta-amyloid (Aß) synaptotoxicity and cognitive dysfunction in mouse models of Alzheimer disease are mediated by a PrP(C)-dependent pathway. Here we review how this pathway converges with proinflammatory cytokine signaling to activate membrane NADPH oxidase (NOX) and generate reactive oxygen species (ROS) leading to dynamic remodeling of the actin cytoskeleton. The NOX signaling pathway may also be integrated with those of other transmembrane receptors clustered in PrP(C)-enriched membrane domains. Such a signal convergence along the PrP(C)-NOX axis could explain the relevance of PrP(C) in a broad spectrum of neurodegenerative disorders, including neuroinflammatory-mediated alterations in synaptic function following traumatic brain injury. PrP(C) overexpression alone activates NOX and generates a local increase in ROS that initiates cofilin activation and formation of cofilin-saturated actin bundles (rods). Rods sequester cofilin from synaptic regions where it is required for plasticity associated with learning and memory. Rods can also interrupt vesicular transport by occluding the neurite within which they form. Through either or both mechanisms, rods may directly mediate the synaptic dysfunction that accompanies various neurodegenerative disorders.


Assuntos
Peptídeos beta-Amiloides/química , Cofilina 1/metabolismo , Neurônios/metabolismo , Príons/química , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Epitopos/química , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Transgênicos , NADPH Oxidases/metabolismo , Doenças Neurodegenerativas/metabolismo , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
12.
PLoS One ; 9(4): e95995, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24760020

RESUMO

Neurites of neurons under acute or chronic stress form bundles of filaments (rods) containing 1∶1 cofilin∶actin, which impair transport and synaptic function. Rods contain disulfide cross-linked cofilin and are induced by treatments resulting in oxidative stress. Rods form rapidly (5-30 min) in >80% of cultured hippocampal or cortical neurons treated with excitotoxic levels of glutamate or energy depleted (hypoxia/ischemia or mitochondrial inhibitors). In contrast, slow rod formation (50% of maximum response in ∼6 h) occurs in a subpopulation (∼20%) of hippocampal neurons upon exposure to soluble human amyloid-ß dimer/trimer (Aßd/t) at subnanomolar concentrations. Here we show that proinflammatory cytokines (TNFα, IL-1ß, IL-6) also induce rods at the same rate and within the same neuronal population as Aßd/t. Neurons from prion (PrP(C))-null mice form rods in response to glutamate or antimycin A, but not in response to proinflammatory cytokines or Aßd/t. Two pathways inducing rod formation were confirmed by demonstrating that NADPH-oxidase (NOX) activity is required for prion-dependent rod formation, but not for rods induced by glutamate or energy depletion. Surprisingly, overexpression of PrP(C) is by itself sufficient to induce rods in over 40% of hippocampal neurons through the NOX-dependent pathway. Persistence of PrP(C)-dependent rods requires the continuous activity of NOX. Removing inducers or inhibiting NOX activity in cells containing PrP(C)-dependent rods causes rod disappearance with a half-life of about 36 min. Cofilin-actin rods provide a mechanism for synapse loss bridging the amyloid and cytokine hypotheses for Alzheimer disease, and may explain how functionally diverse Aß-binding membrane proteins induce synaptic dysfunction.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Neuritos/metabolismo , Proteínas PrPC/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Animais , Células Cultivadas , Dactinomicina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/farmacologia , Humanos , Inflamação/metabolismo , Camundongos , NADPH Oxidases/metabolismo , Proteínas PrPC/genética , Ratos , Transmissão Sináptica/efeitos dos fármacos
13.
PLoS One ; 8(12): e83609, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24391794

RESUMO

Filament bundles (rods) of cofilin and actin (1:1) form in neurites of stressed neurons where they inhibit synaptic function. Live-cell imaging of rod formation is hampered by the fact that overexpression of a chimera of wild type cofilin with a fluorescent protein causes formation of spontaneous and persistent rods, which is exacerbated by the photostress of imaging. The study of rod induction in living cells calls for a rod reporter that does not cause spontaneous rods. From a study in which single cofilin surface residues were mutated, we identified a mutant, cofilinR21Q, which when fused with monomeric Red Fluorescent Protein (mRFP) and expressed several fold above endogenous cofilin, does not induce spontaneous rods even during the photostress of imaging. CofilinR21Q-mRFP only incorporates into rods when they form from endogenous proteins in stressed cells. In neurons, cofilinR21Q-mRFP reports on rods formed from endogenous cofilin and induced by all modes tested thus far. Rods have a half-life of 30-60 min upon removal of the inducer. Vesicle transport in neurites is arrested upon treatments that form rods and recovers as rods disappear. CofilinR21Q-mRFP is a genetically encoded rod reporter that is useful in live cell imaging studies of induced rod formation, including rod dynamics, and kinetics of rod elimination.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Neurônios/metabolismo , Fatores de Despolimerização de Actina/química , Fatores de Despolimerização de Actina/genética , Actinas/química , Actinas/genética , Animais , Linhagem Celular Tumoral , Sistemas Computacionais , Genes Reporter , Células HeLa , Humanos , Células LLC-PK1 , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Modelos Moleculares , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mutagênese Sítio-Dirigida , Estrutura Quaternária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Célula Única , Suínos , Sinapses/metabolismo , Proteína Vermelha Fluorescente
14.
Oxid Med Cell Longev ; 2012: 768101, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22530077

RESUMO

Inflammation and oxidative stress are key to the progressive neuronal degeneration common to chronic pathologies, traumatic injuries, and aging processes in the CNS. The proinflammatory cytokine tumor necrosis factor-alpha (TNF-α) orchestrates cellular stress by stimulating the production and release of neurotoxic mediators including reactive oxygen species (ROS). NADPH oxidases (NOX), ubiquitously expressed in all cells, have recently emerged as pivotal ROS sources in aging and disease. We demonstrated the presence of potent NOX inhibitors in wild Alaska bog blueberries partitioning discretely into a nonpolar fraction with minimal antioxidant capacity and largely devoid of polyphenols. Incubation of SH-SY5Y human neuroblastoma cells with nonpolar blueberry fractions obstructed the coalescing of lipid rafts into large domains disrupting NOX assembly therein and abolishing ROS production characteristic for TNF-α exposure. These findings illuminate nutrition-derived lipid raft modulation as a novel therapeutic approach to blunt inflammatory and oxidative stress in the aging or diseased CNS.


Assuntos
Mirtilos Azuis (Planta)/química , NADPH Oxidases/metabolismo , Neurônios/efeitos dos fármacos , Extratos Vegetais/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Linhagem Celular Tumoral , Humanos , Neurônios/citologia , Neurônios/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/química , Espécies Reativas de Oxigênio/metabolismo
15.
Cell Signal ; 24(6): 1126-33, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22230689

RESUMO

A persistent inflammatory reaction is a hallmark of chronic and acute pathologies in the central nervous system (CNS) and greatly exacerbates neuronal degeneration. The proinflammatory cytokine tumor necrosis factor alpha (TNFα) plays a pivotal role in the initiation and progression of inflammatory processes provoking oxidative stress, eicosanoid biosynthesis, and the production of bioactive lipids. We established in neuronal cells that TNFα exposure dramatically increased Mg(2+)-dependent neutral sphingomyelinase (nSMase) activity thus generating the bioactive lipid mediator ceramide essential for subsequent NADPH oxidase (NOX) activation and oxidative stress. Since many of the pleiotropic effects of ceramide are attributable to its metabolites, we examined whether ceramide kinase (CerK), converting ceramide to ceramide-1-phosphate, is implicated both in NOX activation and enhanced eicosanoid production in neuronal cells. In the present study, we demonstrated that TNFα exposure of human SH-SY5Y neuroblastoma caused a profound increase in CerK activity. Depleting CerK activity using either siRNA or pharmacology completely negated NOX activation and eicosanoid biosynthesis yet, more importantly, rescued neuronal viability in the presence of TNFα. These findings provided evidence for a critical function of ceramide-1-phospate and thus CerK activity in directly linking sphingolipid metabolism to oxidative stress. This vital role of CerK in CNS inflammation could provide a novel therapeutic approach to intervene with the adverse consequences of a progressive CNS inflammation.


Assuntos
NADPH Oxidases/imunologia , Neuroblastoma/imunologia , Fosfotransferases (Aceptor do Grupo Álcool)/imunologia , Fator de Necrose Tumoral alfa/imunologia , Linhagem Celular Tumoral , Ceramidas/imunologia , Ceramidas/metabolismo , Eicosanoides/imunologia , Eicosanoides/metabolismo , Humanos , NADPH Oxidases/metabolismo , Neuroblastoma/metabolismo , Estresse Oxidativo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
16.
Cancer Biol Ther ; 10(11): 1126-36, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20935456

RESUMO

The bioactive sphingolipid ceramide induces oxidative stress by disrupting mitochondrial function and stimulating NADPH oxidase (NOX) activity, both implicated in cell death mechanisms. Many anticancer chemotherapeutics (anthracyclines, Vinca alkaloids, paclitaxel, and fenretinide), as well as physiological stimuli such as tumor necrosis factor α (TNFα), stimulate ceramide accumulation and increase oxidative stress in malignant cells. Consequently, ceramide metabolism in malignant cells and, in particular the up-regulation of glucosylceramide synthase (GCS), has gained considerable interest in contributing to chemoresistance. We hypothesized that increases in GCS activity and thus glucosylceramide, the product of GCS activity, represents an important resistance mechanism in glioblastoma. In our study, we determined that increased GCS activity effectively blocked reactive oxygen species formation by NOX. We further showed, in both glioblastoma and neuroblastoma cells that glucosylceramide directly interfered with NOX assembly, hence delineating a direct resistance mechanism. Collectively, our findings indicated that pharmacological or molecular targeting of GCS, using non-toxic nanoliposome delivery systems, successfully augmented NOX activity, and improved the efficacy of known chemotherapeutic agents.


Assuntos
Glioblastoma/tratamento farmacológico , Glioblastoma/enzimologia , Glucosilceramidas/farmacologia , NADPH Oxidases/antagonistas & inibidores , Neuroblastoma/tratamento farmacológico , Neuroblastoma/enzimologia , Catalase/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/patologia , Glucosilceramidas/metabolismo , Glucosiltransferases/antagonistas & inibidores , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Humanos , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Neuroblastoma/patologia , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
17.
Methods Cell Biol ; 71: 67-87, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12884687

RESUMO

The chick embryo has a long tradition as a model organism in developmental biology as well as embryology. A year-round supply of fertilized eggs, accessibility to all stages of development, and the ease of manipulation of the embryo all contribute to the advantages of investigations using chick embryos. A plethora of culture systems have been developed over the past century allowing to culture intact embryos from as early as 2 days of development. Other culture systems include whole embryo slices, organotypic cultures, tissue explants, and dissociated cultures. Studies utilizing the chick embryo, and in particular spinal motor neurons, were crucial for our present knowledge of the development but also adult physiology, injury, and disease of the nervous system. Extensive studies on spinal motor neurons revealed many molecular mechanisms underlying fundamental events, such as neural induction, axon guidance, programmed cell death, and neuron-target interaction. Cultures of dissociated spinal motor neurons represent one important experimental paradigm. This chapter describes two alternative procedures to establish dissociated spinal motor neuron cultures with virtually no contamination by nonneuronal cells.


Assuntos
Técnicas de Cultura de Células/métodos , Células Cultivadas/citologia , Neurônios Motores/citologia , Medula Espinal/citologia , Animais , Técnicas de Cultura de Células/instrumentação , Separação Celular/instrumentação , Separação Celular/métodos , Células Cultivadas/fisiologia , Centrifugação com Gradiente de Concentração/métodos , Embrião de Galinha , Meios de Cultura/farmacologia , Dissecação/instrumentação , Dissecação/métodos , Neurônios Motores/fisiologia , Medula Espinal/embriologia , Medula Espinal/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa