Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Hum Genet ; 141(3-4): 363-382, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34232383

RESUMEN

Stereocilia protrude up to 100 µm from the apical surface of vertebrate inner ear hair cells and are packed with cross-linked filamentous actin (F-actin). They function as mechanical switches to convert sound vibration into electrochemical neuronal signals transmitted to the brain. Several genes encode molecular components of stereocilia including actin monomers, actin regulatory and bundling proteins, motor proteins and the proteins of the mechanotransduction complex. A stereocilium F-actin core is a dynamic system, which is continuously being remodeled while maintaining an outwardly stable architecture under the regulation of F-actin barbed-end cappers, severing proteins and crosslinkers. The F-actin cores of stereocilia also provide a pathway for motor proteins to transport cargos including components of tip-link densities, scaffolding proteins and actin regulatory proteins. Deficiencies and mutations of stereocilia components that disturb this "dynamic equilibrium" in stereocilia can induce morphological changes and disrupt mechanotransduction causing sensorineural hearing loss, best studied in mouse and zebrafish models. Currently, at least 23 genes, associated with human syndromic and nonsyndromic hearing loss, encode proteins involved in the development and maintenance of stereocilia F-actin cores. However, it is challenging to predict how variants associated with sensorineural hearing loss segregating in families affect protein function. Here, we review the functions of several molecular components of stereocilia F-actin cores and provide new data from our experimental approach to directly evaluate the pathogenicity and functional impact of reported and novel variants of DIAPH1 in autosomal-dominant DFNA1 hearing loss using single-molecule fluorescence microscopy.


Asunto(s)
Sordera , Pérdida Auditiva Sensorineural , Actinas/genética , Animales , Sordera/genética , Sordera/metabolismo , Forminas , Cabello/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Mecanotransducción Celular/genética , Ratones , Proteínas de Microfilamentos/genética , Estereocilios/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo
2.
Clin Genet ; 101(4): 466-471, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35060117

RESUMEN

Diaphanous-related formin 1 (DIAPH1) is a formin homology F-actin elongating protein encoded by DIAPH1. Homozygous recessive variants resulting in the loss of DIAPH1 function cause seizures, cortical blindness, and microcephaly syndrome (SCBMS), but hearing loss has not been reported. In contrast, dominant variants of human DIAPH1 are associated with DFNA1 non-syndromic sensorineural hearing loss. The deafness phenotype is due partly to abnormal F-actin elongation activity caused by disruption of the DIAPH1 autoinhibitory mechanism. We report an elderly female heterozygous for the c.3145C>T: p.R1049X variant who showed late-onset sensorineural hearing loss in her fifth decade. p.R1049X lacks F-actin elongation activity because this variant truncates one-third of the FH2 domain, which is vital for DIAPH1 dimerization and processive F-actin elongation activity. Concordantly, no increase of F-actin or processive F-actin elongation activity was observed after overexpression of p.R1049X DIAPH1 in HeLa cells or by single-molecule microscopy using Xenopus XTC cells. However, overexpression of the p.R1049X variant impairs formation of cell-cell junctions and mitosis. We speculate that late-onset hearing loss is a long-term consequence of heterozygosity for the recessive p.R1049X variant, a phenotype that may have been overlooked among carriers of other recessive alleles of DIAPH1.


Asunto(s)
Forminas , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Anciano , Femenino , Forminas/genética , Células HeLa , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/genética , Humanos , Fenotipo
3.
Hum Mol Genet ; 28(9): 1530-1547, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30602030

RESUMEN

Epilepsy, deafness, onychodystrophy, osteodystrophy and intellectual disability are associated with a spectrum of mutations of human TBC1D24. The mechanisms underlying TBC1D24-associated disorders and the functions of TBC1D24 are not well understood. Using CRISPR-Cas9 genome editing, we engineered a mouse with a premature translation stop codon equivalent to human S324Tfs*3, a recessive mutation of TBC1D24 associated with early infantile epileptic encephalopathy (EIEE). Homozygous S324Tfs*3 mice have normal auditory and vestibular functions but show an abrupt onset of spontaneous seizures at postnatal day 15 recapitulating human EIEE. The S324Tfs*3 variant is located in an alternatively spliced micro-exon encoding six perfectly conserved amino acids incorporated postnatally into TBC1D24 protein due to a micro-exon utilization switch. During embryonic and early postnatal development, S324Tfs*3 homozygotes produce predominantly the shorter wild-type TBC1D24 protein isoform that omits the micro-exon. S324Tfs*3 homozygotes show an abrupt onset of seizures at P15 that correlates with a developmental switch to utilization of the micro-exon. A mouse deficient for alternative splice factor SRRM3 impairs incorporation of the Tbc1d24 micro-exon. Wild-type Tbc1d24 mRNA is abundantly expressed in the hippocampus using RNAscope in situ hybridization. Immunogold electron microscopy using a TBC1D24-specific antibody revealed that TBC1D24 is associated with clathrin-coated vesicles and synapses of hippocampal neurons, suggesting a crucial role of TBC1D24 in vesicle trafficking important for neuronal signal transmission. This is the first characterization of a mouse model of human TBC1D24-associated EIEE that can now be used to screen for antiepileptogenic drugs ameliorating TBCID24 seizure disorders.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Fenotipo , Espasmos Infantiles/diagnóstico , Espasmos Infantiles/genética , Alelos , Animales , Biomarcadores , Encéfalo/metabolismo , Análisis Mutacional de ADN , Proteínas Activadoras de GTPasa/metabolismo , Expresión Génica , Sitios Genéticos , Humanos , Masculino , Ratones , Neuronas/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
J Med Genet ; 56(12): 818-827, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31473629

RESUMEN

BACKGROUND: Diaphanous-related formin 1 (DIA1), which assembles the unbranched actin microfilament and microtubule cytoskeleton, is encoded by DIAPH1. Constitutive activation by the disruption of autoinhibitory interactions between the N-terminal diaphanous inhibitory domain (DID) and C-terminal diaphanous autoregulatory domain (DAD) dysregulates DIA1, resulting in both hearing loss and blood cell abnormalities. METHODS AND RESULTS: Here, we report the first constitutively active mutant in the DID (p.A265S) of humans with only hearing loss and not blood cell abnormality through whole exome sequencing. The previously reported DAD mutants and our DID mutant (p.A265S) shared the finding of diminished autoinhibitory interaction, abnormally upregulated actin polymerisation activity and increased localisations at the plasma membrane. However, the obvious defect in the DIA1-driven assembly of cytoskeleton 'during cell division' was only from the DAD mutants, not from p.A265S, which did not show any blood cell abnormality. We also evaluated the five DID mutants in the hydrophobic pocket since four of these five additional mutants were predicted to critically disrupt interaction between the DID and DAD. These additional pathogenic DID mutants revealed varying degrees of defect in the DIA1-driven cytoskeleton assembly, including nearly normal phenotype during cell division as well as obvious impaired autoinhibition, again coinciding with our key observation in DIA1 mutant (p.A265S) in the DID. CONCLUSION: Here, we report the first mutant in the DID of humans with only hearing loss. The differential cell biological phenotypes of DIA1 during cell division appear to be potential determinants of the clinical severity of DIAPH1-related cytoskeletopathy in humans.


Asunto(s)
División Celular/genética , Citoesqueleto/genética , Forminas/genética , Pérdida Auditiva/genética , Citoesqueleto de Actina/genética , Citoesqueleto/patología , Femenino , Estudios de Asociación Genética , Pérdida Auditiva/patología , Humanos , Masculino , Microtúbulos/genética , Proteínas Mutantes/genética , Mutación/genética , Dominios Proteicos/genética , Secuenciación del Exoma
5.
BMC Neurosci ; 15: 66, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24884926

RESUMEN

BACKGROUND: Notch signaling plays a crucial role in the fate determination of cochlear progenitor cells, hair cells, and supporting cells in the developing cochlea. Recent studies have demonstrated the temporal activation of Notch signaling in damaged mature cochleae, and have demonstrated the induction of new hair cells by pharmacologically inhibiting Notch signaling. The present study aimed to illustrate the feasibility of pharmacologically inhibiting Notch signaling by using a gamma-secretase inhibitor for treating sensorineural hearing loss. RESULTS: The effect of the sustained local delivery of MDL28170, a gamma-secretase inhibitor, on hearing and hair cell induction was tested in a guinea pig model with noise-induced hearing loss. MDL28170 was directly delivered into the cochlear fluids via a micro-osmotic pump. Drug application was initiated 7 days after noise exposure. Measurements of auditory brainstem responses revealed better hearing in the MDL28170-treated animals than in the vehicle controls. Histological analysis demonstrated a higher number of outer hair cells in the MDL28170-treated cochleae than the vehicle-treated cochleae. CONCLUSION: These findings strongly suggest that local sustained delivery of a gamma-secretase inhibitor into the cochlea could be a novel strategy for treating acute hearing loss that is refractory to conventional treatment.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Dipéptidos/uso terapéutico , Células Ciliadas Auditivas Externas/efectos de los fármacos , Células Ciliadas Auditivas Externas/patología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Provocada por Ruido/terapia , Receptores Notch/metabolismo , Animales , Inhibidores de Cisteína Proteinasa/uso terapéutico , Estudios de Factibilidad , Cobayas , Pérdida Auditiva Provocada por Ruido/diagnóstico , Humanos , Resultado del Tratamiento
6.
Front Physiol ; 15: 1374901, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38562617

RESUMEN

Deleterious variants of more than one hundred genes are associated with hearing loss including MYO3A, MYO6, MYO7A and MYO15A and two conventional myosins MYH9 and MYH14. Variants of MYO7A also manifest as Usher syndrome associated with dysfunction of the retina and vestibule as well as hearing loss. While the functions of MYH9 and MYH14 in the inner ear are debated, MYO3A, MYO6, MYO7A and MYO15A are expressed in inner ear hair cells along with class-I myosin MYO1C and are essential for developing and maintaining functional stereocilia on the apical surface of hair cells. Stereocilia are large, cylindrical, actin-rich protrusions functioning as biological mechanosensors to detect sound, acceleration and posture. The rigidity of stereocilia is sustained by highly crosslinked unidirectionally-oriented F-actin, which also provides a scaffold for various proteins including unconventional myosins and their cargo. Typical myosin molecules consist of an ATPase head motor domain to transmit forces to F-actin, a neck containing IQ-motifs that bind regulatory light chains and a tail region with motifs recognizing partners. Instead of long coiled-coil domains characterizing conventional myosins, the tails of unconventional myosins have various motifs to anchor or transport proteins and phospholipids along the F-actin core of a stereocilium. For these myosins, decades of studies have elucidated their biochemical properties, interacting partners in hair cells and variants associated with hearing loss. However, less is known about how myosins traffic in a stereocilium using their motor function, and how each variant correlates with a clinical condition including the severity and onset of hearing loss, mode of inheritance and presence of symptoms other than hearing loss. Here, we cover the domain structures and functions of myosins associated with hearing loss together with advances, open questions about trafficking of myosins in stereocilia and correlations between hundreds of variants in myosins annotated in ClinVar and the corresponding deafness phenotypes.

7.
Res Sq ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38826223

RESUMEN

Stereocilia are unidirectional F-actin-based cylindrical protrusions on the apical surface of inner ear hair cells and function as biological mechanosensors of sound and acceleration. Development of functional stereocilia requires motor activities of unconventional myosins to transport proteins necessary for elongating the F-actin cores and to assemble the mechanoelectrical transduction (MET) channel complex. However, how each myosin localizes in stereocilia using the energy from ATP hydrolysis is only partially understood. In this study, we develop a methodology for live-cell single-molecule fluorescence microscopy of organelles protruding from the apical surface using a dual-view light-sheet microscope, diSPIM. We demonstrate that MYO7A, a component of the MET machinery, traffics as a dimer in stereocilia. Movements of MYO7A are restricted when scaffolded by the plasma membrane and F-actin as mediated by MYO7A's interacting partners. Here, we discuss the technical details of our methodology and its future applications including analyses of cargo transportation in various organelles.

8.
bioRxiv ; 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38766013

RESUMEN

Stereocilia are unidirectional F-actin-based cylindrical protrusions on the apical surface of inner ear hair cells and function as biological mechanosensors of sound and acceleration. Development of functional stereocilia requires motor activities of unconventional myosins to transport proteins necessary for elongating the F-actin cores and to assemble the mechanoelectrical transduction (MET) channel complex. However, how each myosin localizes in stereocilia using the energy from ATP hydrolysis is only partially understood. In this study, we develop a methodology for live-cell single-molecule fluorescence microscopy of organelles protruding from the apical surface using a dual-view light-sheet microscope, diSPIM. We demonstrate that MYO7A, a component of the MET machinery, traffics as a dimer in stereocilia. Movements of MYO7A are restricted when scaffolded by the plasma membrane and F-actin as mediated by MYO7A's interacting partners. Here, we discuss the technical details of our methodology and its future applications including analyses of cargo transportation in various organelles.

9.
J Cell Biol ; 175(6): 947-55, 2006 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-17178911

RESUMEN

Actin forms the dendritic nucleation network and undergoes rapid polymerization-depolymerization cycles in lamellipodia. To elucidate the mechanism of actin disassembly, we characterized molecular kinetics of the major filament end-binding proteins Arp2/3 complex and capping protein (CP) using single-molecule speckle microscopy. We have determined the dissociation rates of Arp2/3 and CP as 0.048 and 0.58 s(-1), respectively, in lamellipodia of live XTC fibroblasts. This CP dissociation rate is three orders of magnitude faster than in vitro. CP dissociates slower from actin stress fibers than from the lamellipodial actin network, suggesting that CP dissociation correlates with actin filament dynamics. We found that jasplakinolide, an actin depolymerization inhibitor, rapidly blocked the fast CP dissociation in cells. Consistently, the coexpression of LIM kinase prolonged CP speckle lifetime in lamellipodia. These results suggest that cofilin-mediated actin disassembly triggers CP dissociation from actin filaments. We predict that filament severing and end-to-end annealing might take place fairly frequently in the dendritic nucleation actin arrays.


Asunto(s)
Proteínas de Capping de la Actina/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas de Capping de la Actina/genética , Proteína 2 Relacionada con la Actina/metabolismo , Proteína 3 Relacionada con la Actina/metabolismo , Animales , Línea Celular , Cofilina 1/metabolismo , Proteínas del Citoesqueleto , Citoesqueleto/metabolismo , Dimerización , Fibroblastos/citología , Fibroblastos/metabolismo , Cinética , Quinasas Lim , Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Seudópodos/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-21701246

RESUMEN

Radiation-induced sarcoma usually develops after an interval of more than 10 years from the completion of radiation therapy to the diagnosis of secondary sarcoma. However, the theory of radiation-induced transformation does not rule out postirradiation sarcomas with a short latency period. We experienced the case of a patient with postirradiation leiomyosarcoma of the tongue, which occurred 19 months after he had received chemoradiotherapy. Besides the short latency period, a pseudotumor stage developed between the time of radiation exposure and the development of leiomyosarcoma. In this article, we also describe an immunohistochemical approach to diagnose leiomyosarcoma and the efficacy of a gemcitabine and docetaxel regimen.


Asunto(s)
Granuloma de Células Plasmáticas/radioterapia , Neoplasias Inducidas por Radiación/etiología , Sarcoma/etiología , Neoplasias de la Lengua/etiología , Biopsia , Diagnóstico Diferencial , Granuloma de Células Plasmáticas/patología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Inducidas por Radiación/diagnóstico , Sarcoma/diagnóstico , Factores de Tiempo , Enfermedades de la Lengua/patología , Enfermedades de la Lengua/radioterapia , Neoplasias de la Lengua/diagnóstico
11.
STAR Protoc ; 2(4): 100967, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34841279

RESUMEN

Fast-dissociating, highly specific monoclonal antibodies (FDSAs) are single-molecule imaging probes useful for many biological assays including consecutive, multiplexable super-resolution microscopy. We developed a screening assay to characterize the kinetics of antibody-antigen interactions using single-molecule microscopy and established a pipeline to identify FDSAs from thousands of monoclonal candidates. Provided here are detailed protocols to prepare multi-well glass-bottom plates necessary for our assay to identify hybridoma clones secreting FDSAs. Synthesis of fluorescently labeled Fab fragments (Fab probes) from FDSAs is also described. For complete details on the use and execution of this protocol, please refer to Miyoshi et al. (2021).


Asunto(s)
Anticuerpos Monoclonales , Colorantes Fluorescentes , Microscopía Fluorescente/métodos , Imagen Individual de Molécula/métodos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Hibridomas/química , Hibridomas/metabolismo , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos BALB C
12.
Cell Rep ; 34(5): 108708, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33535030

RESUMEN

Fast-dissociating, specific antibodies are single-molecule imaging probes that transiently interact with their targets and are used in biological applications including image reconstruction by integrating exchangeable single-molecule localization (IRIS), a multiplexable super-resolution microscopy technique. Here, we introduce a semi-automated screen based on single-molecule total internal reflection fluorescence (TIRF) microscopy of antibody-antigen binding, which allows for identification of fast-dissociating monoclonal antibodies directly from thousands of hybridoma cultures. We develop monoclonal antibodies against three epitope tags (FLAG-tag, S-tag, and V5-tag) and two F-actin crosslinking proteins (plastin and espin). Specific antibodies show fast dissociation with half-lives ranging from 0.98 to 2.2 s. Unexpectedly, fast-dissociating yet specific antibodies are not so rare. A combination of fluorescently labeled Fab probes synthesized from these antibodies and light-sheet microscopy, such as dual-view inverted selective plane illumination microscopy (diSPIM), reveal rapid turnover of espin within long-lived F-actin cores of inner-ear sensory hair cell stereocilia, demonstrating that fast-dissociating specific antibodies can identify novel biological phenomena.


Asunto(s)
Anticuerpos/metabolismo , Hibridomas/metabolismo , Microscopía/métodos , Imagen Individual de Molécula/métodos , Animales , Técnicas de Cultivo de Célula , Humanos , Ratones
13.
J Assoc Res Otolaryngol ; 18(2): 247-261, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28004290

RESUMEN

Aquaporins (AQPs) are a family of small membrane proteins that transport water molecules across the plasma membrane along the osmotic gradient. Mammals express 13 subtypes of AQPs, including the recently reported "subcellular AQPs", AQP11 and 12. Each organ expresses specific subsets of AQP subtypes, and in the inner ear, AQPs are essential for the establishment and maintenance of two distinct fluids, endolymph and perilymph. To evaluate the contribution of AQPs during the establishment of inner ear function, we used quantitative reverse transcription polymerase chain reaction to quantify the expression levels of all known AQPs during the entire development and maturation of the inner ear. Using systematic and longitudinal quantification, we found that AQP11 was majorly and constantly expressed in the inner ear, and that the expression levels of several AQPs follow characteristic longitudinal patterns: increasing (Aqp0, 1, and 9), decreasing (Aqp6, 8, and 12), and peak of expression on E18 (Aqp2, 5, and 7). In particular, the expression level of Aqp9 increased by 70-fold during P3-P21. We also performed in situ hybridization of Aqp11, and determined the unique localization of Aqp11 in the outer hair cells. Immunohistochemistry of AQP9 revealed its localization in the supporting cells inside the organ of Corti, and in the root cells. The emergence of AQP9 expression in these cells was during P3-P21, which was coincident with the marked increase of its expression level. Combining these quantification and localization data, we discuss the possible contributions of these AQPs to inner ear function.


Asunto(s)
Acuaporinas/metabolismo , Oído Interno/metabolismo , Animales , Oído Interno/embriología , Oído Interno/crecimiento & desarrollo , Ratones Endogámicos C57BL
14.
EMBO Mol Med ; 8(11): 1310-1324, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27707755

RESUMEN

DIAPH1 encodes human DIA1, a formin protein that elongates unbranched actin. The c.3634+1G>T DIAPH1 mutation causes autosomal dominant nonsyndromic sensorineural hearing loss, DFNA1, characterized by progressive deafness starting in childhood. The mutation occurs near the C-terminus of the diaphanous autoregulatory domain (DAD) of DIA1, which interacts with its N-terminal diaphanous inhibitory domain (DID), and may engender constitutive activation of DIA1. However, the underlying pathogenesis that causes DFNA1 is unclear. We describe a novel patient-derived DIAPH1 mutation (c.3610C>T) in two unrelated families, which results in early termination prior to a basic amino acid motif (RRKR1204-1207) at the DAD C-terminus. The mutant DIA1(R1204X) disrupted the autoinhibitory DID-DAD interaction and was constitutively active. This unscheduled activity caused increased rates of directional actin polymerization movement and induced formation of elongated microvilli. Mice expressing FLAG-tagged DIA1(R1204X) experienced progressive deafness and hair cell loss at the basal turn and had various morphological abnormalities in stereocilia (short, fused, elongated, sparse). Thus, the basic region of the DAD mediates DIA1 autoinhibition; disruption of the DID-DAD interaction and consequent activation of DIA1(R1204X) causes DFNA1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Regulación de la Expresión Génica , Pérdida Auditiva Sensorineural/genética , Animales , Niño , Modelos Animales de Enfermedad , Femenino , Forminas , Pérdida Auditiva Sensorineural/patología , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Análisis de Secuencia de ADN
15.
Cytoskeleton (Hoboken) ; 70(4): 179-90, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23341338

RESUMEN

Actin forms a polarized filament that grows at the barbed end and shrinks at the pointed end. This phenomenon known as "treadmilling" is believed to govern actin filament turnover. However, in the cell, whether actin turnover proceeds by treadmilling or by other reactions, including filament severing, is a debatable issue. Our previous fluorescence single-molecule speckle (SiMS) analysis has yielded data about the lifetime distribution of F-actin, the uncapping kinetics of both the barbed and pointed ends of the filaments and the elongation rate of the barbed end in lamellipodia. Given these parameters, we estimated the rate of disassembly of the pointed end required to achieve the observed fast actin turnover under the assumption of exclusive filament treadmilling. We derived a method for calculating the lifetime of an individual F-actin subunit at a given position in the Arp2/3 complex-nucleated filament. Extension of this derivation revealed that in the absence of disassembly in the other portions of the filaments, at least 100-fold acceleration of the in vitro pointed end disassembly rate is required to achieve observed F-actin lifetime distribution in lamellipodia. It is, therefore, unlikely that treadmilling solely accounts for the actin filament turnover in vivo. Accumulating evidence obtained by SiMS analysis implies a non-treadmilling actin turnover mechanism in which a substantial amount of F-actin might disassemble near the barbed end of the filament.


Asunto(s)
Actinas/metabolismo , Modelos Biológicos , Seudópodos/metabolismo , Animales , Movimiento Celular/fisiología , Humanos , Proteínas de Microfilamentos/metabolismo , Xenopus laevis
17.
PLoS One ; 4(3): e4921, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19290054

RESUMEN

The mechanism of lamellipod actin turnover is still under debate. To clarify the intracellular behavior of the recently-identified actin disruption mechanism, we examined kinetics of AIP1 using fluorescent single-molecule speckle microscopy. AIP1 is thought to cap cofilin-generated actin barbed ends. Here we demonstrate a reduction in actin-associated AIP1 in lamellipodia of cells overexpressing LIM-kinase. Moreover, actin-associated AIP1 was rapidly abolished by jasplakinolide, which concurrently blocked the F-actin-cofilin interaction. Jasplakinolide also slowed dissociation of AIP1, which is analogous to the effect of this drug on capping protein. These findings provide in vivo evidence of the association of AIP1 with barbed ends generated by cofilin-catalyzed filament disruption. Single-molecule observation found distribution of F-actin-associated AIP1 throughout lamellipodia, and revealed even faster dissociation of AIP1 than capping protein. The estimated overall AIP1-associated actin disruption rate, 1.8 microM/s, was one order of magnitude faster than Arp2/3 complex-catalyzed actin nucleation in lamellipodia. This rate does not suffice the filament severing rate predicted in our previous high frequency filament severing-annealing hypothesis. Our data together with recent biochemical studies imply barbed end-preferred frequent filament disruption. Frequent generation of AIP1-associated barbed ends and subsequent release of AIP1 may be the mechanism that facilitates previously observed ubiquitous actin polymerization throughout lamellipodia.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Actinas/metabolismo , Proteínas de Microfilamentos/metabolismo , Seudópodos/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Biocatálisis , Depsipéptidos/farmacología , Microscopía Fluorescente , Sondas Moleculares
18.
Science ; 303(5666): 2007-10, 2004 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-15044801

RESUMEN

mDia1, a Rho effector, belongs to the Formin family of proteins, which shares the conserved tandem FH1-FH2 unit structure. Formins including mDia1 accelerate actin nucleation while interacting with actin filament fast-growing ends. Here our single-molecule imaging revealed fast directional movement of mDia1 FH1-FH2 for tens of microns in living cells. The movement of mDia1 FH1-FH2 was blocked by actin-perturbing drugs, and the speed of mDia1 FH1-FH2 movement appeared to correlate with actin elongation rates. In vitro, mDia1 FH1-FH2 associated persistently with the growing actin barbed end. mDia1 probably moves processively along the growing end of actin filaments in cells, and Formins may be a molecular motility machinery that is independent from motor proteins.


Asunto(s)
Citoesqueleto de Actina/fisiología , Actinas/metabolismo , Proteínas Portadoras/fisiología , Depsipéptidos , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/ultraestructura , Animales , Biopolímeros , Compuestos Bicíclicos Heterocíclicos con Puentes/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Citocalasina D/metabolismo , Citocalasina D/farmacología , Forminas , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/fisiología , Movimiento , Mutación , Miosinas/fisiología , Péptidos Cíclicos/farmacología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Tiazoles/metabolismo , Tiazoles/farmacología , Tiazolidinas , Xenopus , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoA/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA