Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 295(49): 16713-16731, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-32978259

RESUMEN

The actin cytoskeleton is of profound importance to cell shape, division, and intracellular force generation. Profilins bind to globular (G-)actin and regulate actin filament formation. Although profilins are well-established actin regulators, the distinct roles of the dominant profilin, profilin 1 (PFN1), versus the less abundant profilin 2 (PFN2) remain enigmatic. In this study, we use interaction proteomics to discover that PFN2 is an interaction partner of the actin N-terminal acetyltransferase NAA80, and further confirm this by analytical ultracentrifugation. Enzyme assays with NAA80 and different profilins demonstrate that PFN2 binding specifically increases the intrinsic catalytic activity of NAA80. NAA80 binds PFN2 through a proline-rich loop, deletion of which abrogates PFN2 binding. Small-angle X-ray scattering shows that NAA80, actin, and PFN2 form a ternary complex and that NAA80 has partly disordered regions in the N-terminus and the proline-rich loop, the latter of which is partly ordered upon PFN2 binding. Furthermore, binding of PFN2 to NAA80 via the proline-rich loop promotes binding between the globular domains of actin and NAA80, and thus acetylation of actin. However, the majority of cellular NAA80 is stably bound to PFN2 and not to actin, and we propose that this complex acetylates G-actin before it is incorporated into filaments. In conclusion, we reveal a functionally specific role of PFN2 as a stable interactor and regulator of the actin N-terminal acetyltransferase NAA80, and establish the modus operandi for NAA80-mediated actin N-terminal acetylation, a modification with a major impact on cytoskeletal dynamics.


Asunto(s)
Acetiltransferasas/metabolismo , Actinas/metabolismo , Profilinas/metabolismo , Acetilación , Acetiltransferasas/química , Acetiltransferasas/genética , Citoesqueleto de Actina/metabolismo , Actinas/química , Animales , Biocatálisis , Línea Celular , Humanos , Profilinas/química , Profilinas/deficiencia , Profilinas/genética , Unión Proteica , Dominios Proteicos , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Dispersión del Ángulo Pequeño , Ultracentrifugación , Difracción de Rayos X
2.
Mol Biol Cell ; 31(19): 2107-2114, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32614646

RESUMEN

The actin cytoskeleton plays a variety of roles in eukaryotic cell physiology, ranging from cell polarity and migration to cytokinesis. Key to the function of the actin cytoskeleton is the mechanisms that control its assembly, stability, and turnover. Through genetic analyses in Schizosaccharomyces pombe, we found that myo2-S1 (myo2-G515D), a Myosin II mutant allele, was capable of rescuing lethality caused by partial defects in actin nucleation/stability caused, for example, through compromised function of the actin-binding protein Cdc3-profilin. The mutation in myo2-S1 affects the activation loop of Myosin II, which is involved in physical interaction with subdomain 1 of actin and in stimulating the ATPase activity of Myosin. Consistently, actomyosin rings in myo2-S1 cell ghosts were unstable and severely compromised in contraction on ATP addition. These studies strongly suggest a role for Myo2 in actin cytoskeletal disassembly and turnover in vivo, and that compromise of this activity leads to genetic suppression of mutants defective in actin filament assembly/stability at the division site.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Profilinas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Supresión Genética , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Regulación Fúngica de la Expresión Génica , Mutación , Profilinas/deficiencia , Profilinas/genética , Schizosaccharomyces/genética , Schizosaccharomyces/fisiología , Proteínas de Schizosaccharomyces pombe/genética
3.
Br J Cancer ; 119(9): 1106-1117, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30318519

RESUMEN

BACKGROUND: Adhesion-mediated activation of FAK/ERK signalling pathway, enabled by the formation of filopodial protrusions (FLP), has been shown to be an important event for triggering of dormancy-to-proliferation switch and metastatic outgrowth of breast cancer cells (BCC). We studied the role of actin-binding protein profilin1 (Pfn1) in these processes. METHODS: Quantitative immunohistochemistry (IHC) of BC tissue microarray (TMA) and survival analyses of curated transcriptome datasets of BC patients were performed to examine Pfn1's association with certain clinicopathological features. FLP formation and single cell outgrowth of BCC were assessed using a 3D matrigel culture that accurately predicts dormant vs metastatic outgrowth phenotypes of BCC in certain microenvironment. Gene expression studies were performed to identify potential biological pathways that are perturbed under Pfn1-depleted condition. RESULTS: Lower Pfn1 expression is correlated with lower nuclear grade of breast tumours and longer relapse-free survival of BC patients. Pfn1 depletion leads to defects in FLP and outgrowth of BCC but without impairing either FAK or ERK activation. Guided by transcriptome analyses, we further showed that Pfn1 depletion is associated with prominent SMAD3 upregulation. Although knockdown and overexpression experiments revealed that SMAD3 has an inhibitory effect on the outgrowth of breast cancer cells, SMAD3 knockdown alone was not sufficient to enhance the outgrowth potential of Pfn1-depleted BCC suggesting that other proliferation-regulatory pathways in conjunction with SMAD3 upregulation may underlie the outgrowth-deficient phenotype of BCC cells upon depletion of Pfn1. CONCLUSION: Overall, these data suggest that Pfn1 may be a novel biomarker for BC recurrence and a possible target to reduce metastatic outgrowth of BCC.


Asunto(s)
Neoplasias de la Mama/patología , Técnicas de Cultivo de Célula/métodos , Profilinas/deficiencia , Proteína smad3/genética , Regulación hacia Arriba , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Clasificación del Tumor , Pronóstico , Transducción de Señal , Análisis de Supervivencia , Análisis de Matrices Tisulares , Microambiente Tumoral
4.
J Cell Physiol ; 233(1): 259-268, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28233307

RESUMEN

Osteocytes are the most abundant cells in bone and regulate bone metabolism in coordination with osteoblasts and osteoclasts. However, the molecules that control osteocytes are still incompletely understood. Profilin1 is an actin-binding protein that is involved in actin polymerization. Osteocytes possess characteristic dendritic process formed based on actin cytoskeleton. Here, we examined the expression of profilin1 and its function in osteocytes. Profilin1 mRNA was expressed in osteocytic MLO-Y4 cells and its levels were gradually increased along with the time in culture. With regard to functional aspect, knockdown of profilin1 by siRNA enhanced BMP-induced increase in alkaline phosphatase expression levels in MLO-Y4 cells. Profilin1 knockdown suppressed the levels of dendritic processes and migration of MLO-Y4 cells. Since aging causes an increase in ROS in the body, we further examined the effects of hydrogen peroxide on the expression of profilin1. Hydrogen peroxide treatment increased the levels of profilin1 mRNA in MLO-Y4 cells in contrast to the decline in alkaline phosphatase. Profilin1 was expressed not only in MLO-Y4cells but also in the primary cultures of osteocytes. Importantly, profilin1 mRNA levels in primary cultures of osteocytes were higher than those in primary cultures of osteoblasts. To examine in vivo role of profilin1 in osteocytes, profilin1 was conditionally knocked out by using DMP1-cre and profilin1 floxed mice. This conditional deletion of profilin1 specifically in osteocytes resulted in reduction in the levels of bone volume and bone mineral density. These data indicate that profilin1 is expressed in osteocytes and regulates cell shape, migration and bone mass.


Asunto(s)
Movimiento Celular , Forma de la Célula , Fémur/metabolismo , Osteocitos/metabolismo , Profilinas/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Densidad Ósea , Remodelación Ósea , Línea Celular , Movimiento Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Regulación de la Expresión Génica , Genotipo , Peróxido de Hidrógeno/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocitos/efectos de los fármacos , Fenotipo , Cultivo Primario de Células , Profilinas/deficiencia , Profilinas/genética , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Microtomografía por Rayos X
5.
Nat Commun ; 5: 4746, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25187265

RESUMEN

Wiskott-Aldrich syndrome (WAS) is caused by mutations in the WAS gene and is characterized by immunodeficiency, eczema and microthrombocytopenia. The molecular link between WAS mutations and microthrombocytopenia is unknown. Profilin1 (Pfn1) is a key actin-regulating protein that, besides actin, interacts with phosphoinositides and multiple proline-rich proteins, including the WAS protein (WASp)/WASp-interacting protein (WIP) complex. Here we report that mice with a megakaryocyte/platelet-specific Pfn1 deficiency display microthrombocytopenia due to accelerated turnover of platelets and premature platelet release into the bone marrow. Both Pfn1-null mouse platelets and platelets isolated from WAS patients contained abnormally organized and hyperstable microtubules. These results reveal an unexpected function of Pfn1 as a regulator of microtubule organization and point to a previously unrecognized mechanism underlying the platelet formation defect in WAS patients.


Asunto(s)
Plaquetas/metabolismo , Megacariocitos/metabolismo , Microtúbulos/metabolismo , Profilinas/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/metabolismo , Adolescente , Animales , Plaquetas/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Niño , Preescolar , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Hematopoyesis , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Megacariocitos/patología , Ratones , Microtúbulos/patología , Mutación , Profilinas/genética , Transducción de Señal , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/patología , Proteína del Síndrome de Wiskott-Aldrich/genética
6.
PLoS One ; 7(1): e30068, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253883

RESUMEN

Profilins are important regulators of actin dynamics and have been implicated in activity-dependent morphological changes of dendritic spines and synaptic plasticity. Recently, defective presynaptic excitability and neurotransmitter release of glutamatergic synapses were described for profilin2-deficient mice. Both dendritic spine morphology and synaptic plasticity were fully preserved in these mutants, bringing forward the hypothesis that profilin1 is mainly involved in postsynaptic mechanisms, complementary to the presynaptic role of profilin2. To test the hypothesis and to elucidate the synaptic function of profilin1, we here specifically deleted profilin1 in neurons of the adult forebrain by using conditional knockout mice on a CaMKII-cre-expressing background. Analysis of Golgi-stained hippocampal pyramidal cells and electron micrographs from the CA1 stratum radiatum revealed normal synapse density, spine morphology, and synapse ultrastructure in the absence of profilin1. Moreover, electrophysiological recordings showed that basal synaptic transmission, presynaptic physiology, as well as postsynaptic plasticity were unchanged in profilin1 mutants. Hence, loss of profilin1 had no adverse effects on the morphology and function of excitatory synapses. Our data are in agreement with two different scenarios: i) profilins are not relevant for actin regulation in postsynaptic structures, activity-dependent morphological changes of dendritic spines, and synaptic plasticity or ii) profilin1 and profilin2 have overlapping functions particularly in the postsynaptic compartment. Future analysis of double mutant mice will ultimately unravel whether profilins are relevant for dendritic spine morphology and synaptic plasticity.


Asunto(s)
Profilinas/deficiencia , Sinapsis/fisiología , Animales , Región CA1 Hipocampal/fisiología , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Eliminación de Gen , Ratones , Ratones Transgénicos , Plasticidad Neuronal/fisiología , Especificidad de Órganos , Profilinas/metabolismo , Prosencéfalo/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura
7.
Exp Cell Res ; 315(17): 2963-73, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19607826

RESUMEN

The objective of the present study was to evaluate how different ligand interactions of profilin-1 (Pfn1), an actin-binding protein that is upregulated during capillary morphogenesis of vascular endothelial cells (VEC), contribute to migration and capillary forming ability of VEC. We adopted a knockdown-knockin experimental system to stably express either fully functional form or mutants of Pfn1 that are impaired in binding to two of its major ligands, actin (H119E mutant) and proteins containing polyproline domains (H133S mutant), in a human dermal microvascular cell line (HmVEC) against near-null endogenous Pfn1 background. We found that silencing endogenous Pfn1 expression in HmVEC leads to slower random migration, reduced velocity of membrane protrusion and a significant impairment in matrigel-induced cord formation. Only re-expression of fully functional but not any of the two ligand-binding deficient mutants of Pfn1 rescues the above defects. We further show that loss of Pfn1 expression in VEC inhibits three-dimensional capillary morphogenesis, MMP2 secretion and ECM invasion. VEC invasion through ECM is also inhibited when actin and polyproline interactions of Pfn1 are disrupted. Together, these experimental data demonstrate that Pfn1 regulates VEC migration, invasion and capillary morphogenesis through its interaction with both actin and proline-rich ligands.


Asunto(s)
Capilares/citología , Capilares/fisiología , Movimiento Celular/fisiología , Endotelio Vascular/fisiología , Profilinas/metabolismo , Actinas/metabolismo , Endotelio Vascular/citología , Gelatina/análisis , Silenciador del Gen , Humanos , Morfogénesis/fisiología , Péptidos/metabolismo , Faloidina/análisis , Profilinas/deficiencia , Profilinas/genética , Venas Umbilicales/citología , Venas Umbilicales/crecimiento & desarrollo , Venas Umbilicales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA