Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(12): 105361, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37865312

RESUMEN

In recent years, elegant glycomic and glycoproteomic approaches have revealed an intricate glycosylation profile of mammalian brain with enormous spatial and temporal diversities. Nevertheless, at a cellular level, it is unclear how these post-translational modifications affect various proteins to influence crucial neuronal properties. Here, we have investigated the impact of N-linked glycosylation on neuroligins (NLGNs), a class of cell-adhesion molecules that play instructive roles in synapse organization. We found that endogenous NLGN proteins are differentially glycosylated across several regions of murine brain in a sex-independent but isoform-dependent manner. In both rodent primary neurons derived from brain sections and human neurons differentiated from stem cells, all NLGN variants were highly enriched with multiple N-glycan subtypes, which cumulatively ensured their efficient trafficking to the cell surface. Removal of these N-glycosylation residues only had a moderate effect on NLGNs' stability or expression levels but particularly enhanced their retention at the endoplasmic reticulum. As a result, the glycosylation-deficient NLGNs exhibited considerable impairments in their dendritic distribution and postsynaptic accumulation, which in turn, virtually eliminated their ability to recruit presynaptic terminals and significantly reduced NLGN overexpression-induced assemblies of both glutamatergic and GABAergic synapse structures. Therefore, our results highlight an essential mechanistic contribution of N-linked glycosylations in facilitating the appropriate secretory transport of a major synaptic cell-adhesion molecule and promoting its cellular function in neurons.


Asunto(s)
Neuroliginas , Sinapsis , Animales , Humanos , Ratones , Glicosilación , Neuroliginas/genética , Neuroliginas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sinapsis/metabolismo , Neuronas/metabolismo , Células Cultivadas , Polisacáridos/metabolismo , Transporte de Proteínas/fisiología
2.
Cell Rep ; 43(3): 113914, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38451813

RESUMEN

Stroke, trauma, and neurodegenerative disorders cause loss of neurites (axons and dendrites) in addition to neuronal death. Neurite loss may result directly from a primary insult, secondary to parental neuron death, or secondary to a post-injury inflammatory response. Here, we use lipopolysaccharide and the alarmin S100ß to selectively evaluate neurite loss caused by the inflammatory response. Activation of microglia and infiltrating macrophages by these stimuli causes neurite loss that far exceeds neuronal death, both in vitro and in vivo. Neurite loss is accompanied by the formation of cofilactin rods and aggregates (CARs), which are polymers of cofilin-1 and actin induced by oxidative stress and other factors. Mice deficient in either cofilin-1 or the superoxide-generating enzyme NADPH oxidase-2 show reduced CAR formation, neurite loss, and motor impairment. The findings identify a mechanism by which inflammation leads to neurite loss via CAR formation and highlight the relevance of neurite loss to functional impairment.


Asunto(s)
Neuritas , Enfermedades Neurodegenerativas , Ratones , Animales , Neuronas , Axones , Inflamación
3.
Biomedicines ; 12(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38255199

RESUMEN

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.

4.
Cell Death Dis ; 15(4): 264, 2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38615035

RESUMEN

Cognitive dysfunction and dementia are critical symptoms of Lewy Body dementias (LBD). Specifically, alpha-synuclein (αSyn) accumulation in the hippocampus leading to synaptic dysfunction is linked to cognitive deficits in LBD. Here, we investigated the pathological impact of αSyn on hippocampal neurons. We report that either αSyn overexpression or αSyn pre-formed fibrils (PFFs) treatment triggers the formation of cofilin-actin rods, synapse disruptors, in cultured hippocampal neurons and in the hippocampus of synucleinopathy mouse models and of LBD patients. In vivo, cofilin pathology is present concomitantly with synaptic impairment and cognitive dysfunction. Rods generation prompted by αSyn involves the co-action of the cellular prion protein (PrPC) and the chemokine receptor 5 (CCR5). Importantly, we show that CCR5 inhibition, with a clinically relevant peptide antagonist, reverts dendritic spine impairment promoted by αSyn. Collectively, we detail the cellular and molecular mechanism through which αSyn disrupts hippocampal synaptic structure and we identify CCR5 as a novel therapeutic target to prevent synaptic impairment and cognitive dysfunction in LBD.


Asunto(s)
Trastornos del Conocimiento , Enfermedad por Cuerpos de Lewy , Animales , Ratones , Humanos , alfa-Sinucleína , Espinas Dendríticas , Factores Despolimerizantes de la Actina , Receptores CCR5/genética
5.
J Neurosci ; 32(19): 6670-81, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22573689

RESUMEN

Rod-shaped aggregates ("rods"), containing equimolar actin and the actin dynamizing protein cofilin, appear in neurons following a wide variety of potentially oxidative stress: simulated microischemia, cofilin overexpression, and exposure to peroxide, excess glutamate, or the dimer/trimer forms of amyloid-ß peptide (Aßd/t), the most synaptotoxic Aß species. These rods are initially reversible and neuroprotective, but if they persist in neurites, the synapses degenerate without neurons dying. Herein we report evidence that rod formation depends on the generation of intermolecular disulfide bonds in cofilin. Of four Cys-to-Ala cofilin mutations expressed in rat E18 hippocampal neurons, only the mutant incapable of forming intermolecular bonds (CC39,147AA) has significantly reduced ability to incorporate into rods. Rod regions show unusually high oxidation levels. Rods, isolated from stressed neurons, contain dithiothreitol-sensitive multimeric forms of cofilin, predominantly dimer. Oligomerization of cofilin in cells represents one more mechanism for regulating the actin dynamizing activity of cofilin and probably underlies synaptic loss.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/fisiología , Disulfuros/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Factores Despolimerizantes de la Actina/química , Factores Despolimerizantes de la Actina/genética , Animales , Línea Celular Tumoral , Células Cultivadas , Pollos , Disulfuros/química , Femenino , Humanos , Masculino , Ratones , Enfermedades Neurodegenerativas/genética , Oxidación-Reducción , Multimerización de Proteína/genética , Ratas , Porcinos
6.
BMC Cell Biol ; 14: 45, 2013 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-24093776

RESUMEN

BACKGROUND: ADF/cofilin proteins are key modulators of actin dynamics in metastasis and invasion of cancer cells. Here we focused on the roles of ADF and cofilin-1 individually in the development of polarized migration of rat mammary adenocarcinoma (MTLn3) cells, which express nearly equal amounts of each protein. Small interference RNA (siRNA) technology was used to knockdown (KD) the expression of ADF and cofilin-1 independently. RESULTS: Either ADF KD or cofilin KD caused cell elongation, a reduction in cell area, a decreased ability to form invadopodia, and a decreased percentage of polarized cells after 180 s of epidermal growth factor stimulation. Moreover, ADF KD or cofilin KD increased the rate of cell migration and the time of lamellipodia protrusion but through different mechanisms: lamellipodia protrude more frequently in ADF KD cells and are more persistent in cofilin KD cells. ADF KD cells showed a significant increase in F-actin aggregates, whereas cofilin KD cells showed a significant increase in prominent F-actin bundles and increased cell adhesion. Focal adhesion area and cell adhesion in cofilin KD cells were returned to control levels by expressing exogenous cofilin but not ADF. Return to control rates of cell migration in ADF KD cells was achieved by expression of exogenous ADF but not cofilin, whereas in cofilin KD cells, expression of cofilin efficiently rescued control migration rates. CONCLUSION: Although ADF and cofilin have many redundant functions, each of these isoforms has functional differences that affect F-actin structures, cell adhesion and lamellipodial dynamics, all of which are important determinants of cell migration.


Asunto(s)
Actinas/metabolismo , Adenocarcinoma/metabolismo , Cofilina 1/genética , Destrina/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Animales/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Adhesión Celular/efectos de los fármacos , Movimiento Celular , Cofilina 1/antagonistas & inhibidores , Cofilina 1/metabolismo , Destrina/antagonistas & inhibidores , Destrina/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Femenino , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/metabolismo , Adhesiones Focales/ultraestructura , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Metástasis de la Neoplasia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Transducción de Señal , Células Tumorales Cultivadas
7.
Hum Mol Genet ; 20(10): 1937-51, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21355047

RESUMEN

Huntington's disease (HD) is caused by an expanded CAG tract in the Interesting transcript 15 (IT15) gene encoding the 350 kDa huntingtin protein. Cellular stresses can trigger the release of huntingtin from the endoplasmic reticulum, allowing huntingtin nuclear entry. Here, we show that endogenous, full-length huntingtin localizes to nuclear cofilin-actin rods during stress and is required for the proper stress response involving actin remodeling. Mutant huntingtin induces a dominant, persistent nuclear rod phenotype similar to that described in Alzheimer's disease for cytoplasmic cofilin-actin rods. Using live cell temporal studies, we show that this stress response is similarly impaired when mutant huntingtin is present, or when normal huntingtin levels are reduced. In clinical lymphocyte samples from HD patients, we have quantitatively detected cross-linked complexes of actin and cofilin with complex formation varying in correlation with disease progression. By live cell fluorescence lifetime imaging measurement-Förster resonant energy transfer studies and western blot assays, we quantitatively observed that stress-activated tissue transglutaminase 2 (TG2) is responsible for the actin-cofilin covalent cross-linking observed in HD. These data support a direct role for huntingtin in nuclear actin re-organization, and describe a new pathogenic mechanism for aberrant TG2 enzymatic hyperactivity in neurodegenerative diseases.


Asunto(s)
Actinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Respuesta al Choque Térmico/genética , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Transglutaminasas/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Animales , Línea Celular , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al GTP/genética , Expresión Génica/genética , Calor , Humanos , Proteína Huntingtina , Espacio Intracelular/metabolismo , Linfocitos/metabolismo , Ratones , Modelos Biológicos , Células 3T3 NIH , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transporte de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transglutaminasas/genética
8.
Nat Rev Neurosci ; 9(2): 136-47, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18209731

RESUMEN

Higher-order actin-based networks (actin superstructures) are important for growth-cone motility and guidance. Principles for generating, organizing and remodelling actin superstructures have emerged from recent findings in cell-free systems, non-neuronal cells and growth cones. This Review examines how actin superstructures are initiated de novo at the leading-edge membrane and how the spontaneous organization of actin superstructures is driven by ensembles of actin-binding proteins. How the regulation of actin-binding proteins can affect growth-cone turning and axonal regeneration is also discussed.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Diferenciación Celular/fisiología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Conos de Crecimiento/metabolismo , Proteínas de Microfilamentos/metabolismo , Citoesqueleto de Actina/ultraestructura , Animales , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Movimiento Celular/fisiología , Sistema Nervioso Central/citología , Conos de Crecimiento/ultraestructura , Humanos , Vías Nerviosas/citología , Vías Nerviosas/embriología , Vías Nerviosas/metabolismo , Plasticidad Neuronal/fisiología
9.
Methods Mol Biol ; 2593: 265-281, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36513938

RESUMEN

Fluorescence microscopy of cytoskeletal proteins in situ using immunolabeling, fluorescent reagents, or expression of tagged proteins has been a common practice for decades but often with too little regard for what might not be visualized. This is especially true for assembled filamentous actin (F-actin), for which binding of fluorescently labeled phalloidin is taken as the gold standard for its quantification even though it is well known that F-actin saturated with cofilin (cofilactin) binds neither fluorescently labeled phalloidin nor genetically encoded F-actin reporters, such as LifeAct. Here, using expressed fluorescent cofilactin reporters, we show that cofilactin is the major component of some actin-containing structures in both normal and stressed neurons and present various fixation, permeabilization, and cryo-preservation methods for optimizing its observation.


Asunto(s)
Factores Despolimerizantes de la Actina , Actinas , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Faloidina/metabolismo , Citoesqueleto de Actina/metabolismo , Técnica del Anticuerpo Fluorescente
10.
Biomedicines ; 11(11)2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-38001943

RESUMEN

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.

11.
Mol Microbiol ; 81(4): 851-4, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21762221

RESUMEN

Actin reorganization, mediated by the actin dynamizing protein cofilin, is essential for host cell invasion by the intracellular pathogenic bacterium Listeria monocytogenes. During invasion, the InlB bacterial surface ligands closely interact with host cell Met receptors to induce phagocytosis. In this issue of Molecular Microbiology, Han et al., 2011 clearly demonstrate that phospholipase D (PLD)-dependent production of membrane phosphatidic acid is required for invasion. They further show that the phosphorylated form of cofilin, which is inactive in actin binding, is necessary for the activation of the PLD1 isoform. Although cofilin-independent PLD2 can also mediate internalization, it is a phospho-cofilin-dependent balanced production of phosphatidic acid that is required for optimal Listeria internalization. Cofilin-dependent membrane lipid remodelling has important implications for cofilin function that go well beyond its direct effects on actin.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Interacciones Huésped-Patógeno , Listeria monocytogenes/patogenicidad , Ácidos Fosfatidicos/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Fagocitosis , Fosfolipasa D/metabolismo , Fosforilación , Isoformas de Proteínas/metabolismo
12.
J Cell Biol ; 178(1): 107-19, 2007 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-17606869

RESUMEN

Bone morphogenic proteins (BMPs) are involved in axon pathfinding, but how they guide growth cones remains elusive. In this study, we report that a BMP7 gradient elicits bidirectional turning responses from nerve growth cones by acting through LIM kinase (LIMK) and Slingshot (SSH) phosphatase to regulate actin-depolymerizing factor (ADF)/cofilin-mediated actin dynamics. Xenopus laevis growth cones from 4-8-h cultured neurons are attracted to BMP7 gradients but become repelled by BMP7 after overnight culture. The attraction and repulsion are mediated by LIMK and SSH, respectively, which oppositely regulate the phosphorylation-dependent asymmetric activity of ADF/cofilin to control the actin dynamics and growth cone steering. The attraction to repulsion switching requires the expression of a transient receptor potential (TRP) channel TRPC1 and involves Ca2+ signaling through calcineurin phosphatase for SSH activation and growth cone repulsion. Together, we show that spatial regulation of ADF/cofilin activity controls the directional responses of the growth cone to BMP7, and Ca2+ influx through TRPC tilts the LIMK-SSH balance toward SSH-mediated repulsion.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Cofilina 1/metabolismo , Destrina/metabolismo , Conos de Crecimiento/fisiología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Quinasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Blastómeros/metabolismo , Proteína Morfogenética Ósea 7 , Calcio/metabolismo , Señalización del Calcio , Destrina/genética , Embrión no Mamífero , Ganglios Espinales/citología , Quinasas Lim , Microinyecciones , Modelos Biológicos , Neuronas/citología , Neuronas/metabolismo , ARN Mensajero/administración & dosificación , Canales Catiónicos TRPC/metabolismo , Xenopus/embriología , Proteínas de Xenopus/metabolismo
13.
Exp Cell Res ; 317(3): 249-61, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21036167

RESUMEN

Tropomyosins are believed to function in part by stabilizing actin filaments. However, accumulating evidence suggests that fundamental differences in function exist between tropomyosin isoforms, which contributes to the formation of functionally distinct filament populations. We investigated the functions of the high-molecular-weight isoform Tm3 and examined the molecular properties of Tm3-containing actin filament populations. Overexpression of the Tm3 isoform specifically induced the formation of filopodia and changes in actin solubility. We observed alterations in actin-binding protein recruitment to filaments, co-incident with changes in expression levels, which can account for this functional outcome. Tm3-associated filaments recruit active actin depolymerizing factor and are bundled into filopodia by fascin, which is both up-regulated and preferentially associated with Tm3-containing filaments in the Tm3 overexpressing cells. This study provides further insight into the isoform-specific roles of different tropomyosin isoforms. We conclude that variation in the tropomyosin isoform composition of microfilaments provides a mechanism to generate functionally distinct filament populations.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Microfilamentos/metabolismo , Seudópodos/metabolismo , Tropomiosina/fisiología , Actinas/metabolismo , Animales , Línea Celular , Humanos , Isoformas de Proteínas , Transporte de Proteínas , Ratas , Tropomiosina/genética
14.
J Biol Chem ; 285(8): 5450-60, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20022956

RESUMEN

Cofilin-actin bundles (rods), which form in axons and dendrites of stressed neurons, lead to synaptic dysfunction and may mediate cognitive deficits in dementias. Rods form abundantly in the cytoplasm of non-neuronal cells in response to many treatments that induce rods in neurons. Rods in cell lysates are not stable in detergents or with added calcium. Rods induced by ATP-depletion and released from cells by mechanical lysis were first isolated from two cell lines expressing chimeric actin-depolymerizing factor (ADF)/cofilin fluorescent proteins by differential and equilibrium sedimentation on OptiPrep gradients and then from neuronal and non-neuronal cells expressing only endogenous proteins. Rods contain ADF/cofilin and actin in a 1:1 ratio. Isolated rods are stable in dithiothreitol, EGTA, Ca(2+), and ATP. Cofilin-GFP-containing rods are stable in 500 mM NaCl, whereas rods formed from endogenous proteins are significantly less stable in high salt. Proteomic analysis of rods formed from endogenous proteins identified other potential components whose presence in rods was examined by immunofluorescence staining of cells. Only actin and ADF/cofilin are in rods during all phases of their formation; furthermore, the rapid assembly of rods in vitro from these purified proteins at physiological concentration shows that they are the only proteins necessary for rod formation. Cytoplasmic rod formation is inhibited by cytochalasin D and jasplakinolide. Time lapse imaging of rod formation shows abundant small needle-shaped rods that coalesce over time. Rod filament lengths measured by ultrastructural tomography ranged from 22 to 1480 nm. These results suggest rods form by assembly of cofilin-actin subunits, followed by self-association of ADF/cofilin-saturated F-actin.


Asunto(s)
Factores Despolimerizantes de la Actina/química , Factores Despolimerizantes de la Actina/aislamiento & purificación , Actinas/química , Actinas/aislamiento & purificación , Destrina/química , Destrina/aislamiento & purificación , Complejos Multiproteicos/química , Complejos Multiproteicos/aislamiento & purificación , Factores Despolimerizantes de la Actina/genética , Factores Despolimerizantes de la Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Destrina/genética , Destrina/metabolismo , Células HeLa , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Ratas , Porcinos , Xenopus laevis
15.
Cells ; 10(10)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34685706

RESUMEN

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.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Degeneración Nerviosa/patología , Neuronas/metabolismo , Factores Despolimerizantes de la Actina/química , Secuencia de Aminoácidos , Animales , Humanos , Neuritas/metabolismo , Neurogénesis
16.
PLoS One ; 16(3): e0248309, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33705493

RESUMEN

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.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Hipocampo/metabolismo , NADPH Oxidasas/metabolismo , Neuronas/metabolismo , Proteínas PrPC/metabolismo , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Factores Despolimerizantes de la Actina/genética , Actinas/genética , Animales , Proteína gp120 de Envoltorio del VIH/genética , Infecciones por VIH/genética , VIH-1/genética , Ratones , Ratones Noqueados , NADPH Oxidasas/genética , Estrés Oxidativo/genética , Proteínas PrPC/genética , Receptores CCR5/genética , Receptores CXCR4/genética
17.
J Neurosci ; 29(41): 12994-3005, 2009 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-19828813

RESUMEN

In Alzheimer's disease (AD), rod-like cofilin aggregates (cofilin-actin rods) and thread-like inclusions containing phosphorylated microtubule-associated protein (pMAP) tau form in the brain (neuropil threads), and the extent of their presence correlates with cognitive decline and disease progression. The assembly mechanism of these respective pathological lesions and the relationship between them is poorly understood, yet vital to understanding the causes of sporadic AD. We demonstrate that, during mitochondrial inhibition, activated actin-depolymerizing factor (ADF)/cofilin assemble into rods along processes of cultured primary neurons that recruit pMAP/tau and mimic neuropil threads. Fluorescence resonance energy transfer analysis revealed colocalization of cofilin-GFP (green fluorescent protein) and pMAP in rods, suggesting their close proximity within a cytoskeletal inclusion complex. The relationship between pMAP and cofilin-actin rods was further investigated using actin-modifying drugs and small interfering RNA knockdown of ADF/cofilin in primary neurons. The results suggest that activation of ADF/cofilin and generation of cofilin-actin rods is required for the subsequent recruitment of pMAP into the inclusions. Additionally, we were able to induce the formation of pMAP-positive ADF/cofilin rods by exposing cells to exogenous amyloid-beta (Abeta) peptides. These results reveal a common pathway for pMAP and cofilin accumulation in neuronal processes. The requirement of activated ADF/cofilin for the sequestration of pMAP suggests that neuropil thread structures in the AD brain may be initiated by elevated cofilin activation and F-actin bundling that can be caused by oxidative stress, mitochondrial dysfunction, or Abeta peptides, all suspected initiators of synaptic loss and neurodegeneration in AD.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Neuritas/metabolismo , Neuronas/patología , Proteínas tau/metabolismo , Factores Despolimerizantes de la Actina/genética , Adenosina Trifosfato/farmacología , Enfermedad de Alzheimer/patología , Secuencias de Aminoácidos/fisiología , Péptidos beta-Amiloides/farmacología , Animales , Animales Recién Nacidos , Antimicina A/análogos & derivados , Antimicina A/farmacología , Encéfalo/patología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Células Cultivadas , Embrión de Pollo/citología , Depsipéptidos/farmacología , Inhibidores Enzimáticos/farmacología , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Fluorescentes Verdes/genética , Humanos , Peróxido de Hidrógeno/farmacología , Ionóforos/farmacología , Neuritas/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Oxidantes/farmacología , Fragmentos de Péptidos/farmacología , Fosforilación/fisiología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Serina/metabolismo , Tiazolidinas/farmacología , Transfección/métodos , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo
18.
Cell Motil Cytoskeleton ; 66(8): 635-49, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19479823

RESUMEN

The histopathological hallmarks of Alzheimer disease are the extracellular amyloid plaques, composed principally of the amyloid beta peptide, and the intracellular neurofibrillary tangles, composed of paired helical filaments of the microtubule-associated protein, tau. Other histopathological structures involving actin and the actin-binding protein, cofilin, have more recently been recognized. Here we review new findings about these cytoskeletal pathologies, and, emphasize how plaques, tangles, the actin-containing inclusions and their respective building blocks may contribute to Alzheimer pathogenesis and the primary behavioral symptoms of the disease. Cell Motil. Cytoskeleton, 2009. (c) 2009 Wiley-Liss, Inc.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Citoesqueleto/patología , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Citoesqueleto/metabolismo , Humanos , Microtúbulos/metabolismo
19.
Mol Biol Cell ; 31(21): 2363-2378, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32816614

RESUMEN

Nuclear envelope proteins influence cell cytoarchitecure by poorly understood mechanisms. Here we show that small interfering RNA-mediated silencing of lamin A/C (LMNA) promotes contrasting stress fiber assembly and disassembly in individual cells and within cell populations. We show that LMNA-deficient cells have elevated myosin-II bipolar filament accumulations, irregular formation of actin comet tails and podosome-like adhesions, increased steady state nuclear localization of the mechanosensitive transcription factors MKL1 and YAP, and induced expression of some MKL1/serum response factor-regulated genes such as that encoding myosin-IIA (MYH9). Our studies utilizing live cell imaging and pharmacological inhibition of myosin-II support a mechanism of deregulated myosin-II self-organizing activity at the nexus of divergent actin cytoskeletal aberrations resulting from LMNA loss. In light of our results, we propose a model of how the nucleus, via linkage to the cytoplasmic actomyosin network, may act to control myosin-II contractile behavior through both mechanical and transcriptional feedback mechanisms.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Lamina Tipo A/metabolismo , Miosina Tipo II/metabolismo , Membrana Nuclear/metabolismo , Línea Celular , Línea Celular Tumoral , Regulación de la Expresión Génica , Células HeLa , Humanos , Lamina Tipo A/deficiencia , Miosina Tipo II/genética
20.
Trends Cell Biol ; 12(12): 598-605, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12495849

RESUMEN

ADF/cofilins are key regulators of actin dynamics in normal cells. Recent findings suggest that, under cellular stress, the wild-type proteins might form complexes with actin that can alter cell function. Owing to their rapid formation, these complexes might initiate or aid in the progression of diseases as diverse as Alzheimer's disease and ischemic kidney disease. Although evidence for their involvement in diseases other than Alzheimer's and ischemic kidney disease is tenuous, recent studies suggest that altered production, regulation or localization of these proteins might lead to cognitive impairment, inflammation, infertility, immune deficiencies and other pathophysiological defects.


Asunto(s)
Actinas/metabolismo , Infertilidad/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias/metabolismo , Factores Despolimerizantes de la Actina , Actinas/química , Animales , Quimiotaxis , Destrina , Humanos , Proteínas de Microfilamentos/química , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA