Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Biol Chem ; 300(1): 105516, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38042485

RESUMEN

Class III myosins localize to inner ear hair cell stereocilia and are thought to be crucial for stereocilia length regulation. Mutations within the motor domain of MYO3A that disrupt its intrinsic motor properties have been associated with non-syndromic hearing loss, suggesting that the motor properties of MYO3A are critical for its function within stereocilia. In this study, we investigated the impact of a MYO3A hearing loss mutation, H442N, using both in vitro motor assays and cell biological studies. Our results demonstrate the mutation causes a dramatic increase in intrinsic motor properties, actin-activated ATPase and in vitro actin gliding velocity, as well as an increase in actin protrusion extension velocity. We propose that both "gain of function" and "loss of function" mutations in MYO3A can impair stereocilia length regulation, which is crucial for stereocilia formation during development and normal hearing. Furthermore, we generated chimeric MYO3A constructs that replace the MYO3A motor and neck domain with the motor and neck domain of other myosins. We found that duty ratio, fraction of ATPase cycle myosin is strongly bound to actin, is a critical motor property that dictates the ability to tip localize within filopodia. In addition, in vitro actin gliding velocities correlated extremely well with filopodial extension velocities over a wide range of gliding and extension velocities. Taken together, our data suggest a model in which tip-localized myosin motors exert force that slides the membrane tip-ward, which can combat membrane tension and enhance the actin polymerization rate that ultimately drives protrusion elongation.


Asunto(s)
Actinas , Pérdida Auditiva , Miosina Tipo III , Animales , Actinas/genética , Actinas/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Chlorocebus aethiops , Células COS , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Miosina Tipo III/genética , Miosina Tipo III/metabolismo , Miosinas/genética , Miosinas/metabolismo , Estereocilios , Humanos
2.
Eur J Hum Genet ; 30(1): 13-21, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33953343

RESUMEN

We recently described a novel missense variant [c.2090T>G:p.(Leu697Trp)] in the MYO3A gene, found in two Brazilian families with late-onset autosomal dominant nonsyndromic hearing loss (ADNSHL). Since then, with the objective of evaluating its contribution to ADNSHL in Brazil, the variant was screened in additional 101 pedigrees with probable ADNSHL without conclusive molecular diagnosis. The variant was found in three additional families, explaining 3/101 (~3%) of cases with ADNSHL in our Brazilian pedigree collection. In order to identify the origin of the variant, 21 individuals from the five families were genotyped with a high-density SNP array (~600 K SNPs- Axiom Human Origins; ThermoFisher). The identity by descent (IBD) approach revealed that many pairs of individuals from the different families have a kinship coefficient equivalent to that of second cousins, and all share a minimum haplotype of ~607 kb which includes the c.2090T>G variant suggesting it probably arose in a common ancestor. We inferred that the mutation occurred in a chromosomal segment of European ancestry and the time since the most common ancestor was estimated in 1100 years (CI = 775-1425). This variant was also reported in a Dutch family, which shares a 87,121 bp haplotype with the Brazilian samples, suggesting that Dutch colonists may have brought it to Northeastern Brazil in the 17th century. Therefore, the present study opens new avenues to investigate this variant not only in Brazilians but also in European families with ADNSHL.


Asunto(s)
Frecuencia de los Genes , Pérdida Auditiva Sensorineural/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Brasil , Efecto Fundador , Genes Dominantes , Haplotipos , Migración Humana , Humanos , Mutación Missense , Linaje , Polimorfismo de Nucleótido Simple
3.
Mol Biol Cell ; 33(1): ar5, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34788109

RESUMEN

Class III myosins are actin-based motors proposed to transport cargo to the distal tips of stereocilia in the inner ear hair cells and/or to participate in stereocilia length regulation, which is especially important during development. Mutations in the MYO3A gene are associated with delayed onset deafness. A previous study demonstrated that L697W, a dominant deafness mutation, disrupts MYO3A ATPase and motor properties but does not impair its ability to localize to the tips of actin protrusions. In the current study, we characterized the transient kinetic mechanism of the L697W motor ATPase cycle. Our kinetic analysis demonstrates that the mutation slows the ADP release and ATP hydrolysis steps, which results in a slight reduction in the duty ratio and slows detachment kinetics. Fluorescence recovery after photobleaching (FRAP) of filopodia tip localized L697W and WT MYO3A in COS-7 cells revealed that the mutant does not alter turnover or average intensity at the actin protrusion tips. We demonstrate that the mutation slows filopodia extension velocity in COS-7 cells which correlates with its twofold slower in vitro actin gliding velocity. Overall, this work allowed us to propose a model for how the motor properties of MYO3A are crucial for facilitating actin protrusion length regulation.


Asunto(s)
Sordera/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/genética , Miosina Tipo III/metabolismo , Actinas/metabolismo , Adenosina Trifosfatasas/genética , Animales , Células COS , Chlorocebus aethiops , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Humanos , Cinética , Mutación , Miosinas , Seudópodos/metabolismo
4.
J Biomol Struct Dyn ; 40(21): 10940-10951, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34423747

RESUMEN

Hereditary hearing impairment (HI) is a common disease with the highest incidence among sensory defects. Several genes have been identified to affect stereocilia structure causing HI, including the unconventional myosin3A. Interestingly, we noticed that variants in MYO3A gene have been previously found to cause variable HI onset and severity. Using clinical exome sequencing, we identified a novel pathogenic variant p.(Lys50Arg) in the MYO3A kinase domain (MYO3A-KD). Previous in vitro studies supported its damaging effect as a 'kinase-dead' mutant. We further analyzed this variation through molecular dynamics which predicts that changes in flexibility of MYO3A structure would influence the protein-ATP binding properties. This Lys50Arg mutation segregated with congenital profound non-syndromic HI. To better investigate this variability, we collected previously identified MYO3A-KDs variants, p.(Tyr129Cys), p.(His142Gln) and p.(Pro189Thr), and built both wild type and mutant 3 D MYO3A-KD models to assess their impact on the protein structure and function. Our results suggest that KD mutations could either cause a congenital profound form of HI, when particularly affecting the kinase activity and preventing the auto-phosphorylation of the motor, or a late onset and progressive form, when partially or completely inactivating the MYO3A protein. In conclusion, we report a novel pathogenic variant affecting the ATP-binding site within the MYO3A-KD causing congenital profound HI. Through computational approaches we provide a deeper understanding on the correlation between the effects of MYO3A-KD mutations and the variable hearing phenotypes. To the best of our knowledge this is the first study to correlate mutations' genotypes with the variable phenotypes of DFNB30.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Pérdida Auditiva Sensorineural , Pérdida Auditiva , Miosina Tipo III , Humanos , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Mutación , Adenosina Trifosfato , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética
5.
Hum Mol Genet ; 29(23): 3729-3743, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33078831

RESUMEN

There is scarcity of known gene variants of hearing impairment (HI) in African populations. This knowledge deficit is ultimately affecting the development of genetic diagnoses. We used whole exome sequencing to investigate gene variants, pathways of interactive genes and the fractions of ancestral overderived alleles for 159 HI genes among 18 Cameroonian patients with non-syndromic HI (NSHI) and 129 ethnically matched controls. Pathogenic and likely pathogenic (PLP) variants were found in MYO3A, MYO15A and COL9A3, with a resolution rate of 50% (9/18 patients). The study identified significant genetic differentiation in novel population-specific gene variants at FOXD4L2, DHRS2L6, RPL3L and VTN between HI patients and controls. These gene variants are found in functional/co-expressed interactive networks with other known HI-associated genes and in the same pathways with VTN being a hub protein, that is, focal adhesion pathway and regulation of the actin cytoskeleton (P-values <0.05). The results suggest that these novel population-specific gene variants are possible modifiers of the HI phenotypes. We found a high proportion of ancestral allele versus derived at low HI patients-specific minor allele frequency in the range of 0.0-0.1. The results showed a relatively low pickup rate of PLP variants in known genes in this group of Cameroonian patients with NSHI. In addition, findings may signal an evolutionary enrichment of some variants of HI genes in patients, as the result of polygenic adaptation, and suggest the possibility of multigenic influence on the phenotype of congenital HI, which deserves further investigations.


Asunto(s)
Colágeno Tipo IX/genética , Secuenciación del Exoma/métodos , Pérdida Auditiva/patología , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Miosinas/genética , Adulto , Alelos , Camerún/epidemiología , Estudios de Casos y Controles , Niño , Femenino , Pérdida Auditiva/epidemiología , Pérdida Auditiva/genética , Humanos , Masculino , Fenotipo
6.
Mol Genet Genomic Med ; 8(8): e1343, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32519820

RESUMEN

BACKGROUND: MYO3A, encoding the myosin IIIA protein, is associated with autosomal recessive and autosomal dominant nonsyndromic hearing loss. To date, only two missense variants located in the motor-head domain of MYO3A have been described in autosomal dominant families with progressive, mild-to-profound sensorineural hearing loss. These variants alter the ATPase activity of myosin IIIA. METHODS: Exome sequencing of a proband from a three-generation German family with prelingual, moderate-to-profound, high-frequency hearing loss was performed. Segregation analysis confirmed a dominant inheritance pattern. Regression analysis of mean hearing level thresholds per individual and ear was performed at high-, mid-, and low-frequencies. RESULTS: A novel heterozygous missense variant c.716T>C, p.(Leu239Pro) in the kinase domain of MYO3A was identified that is predicted in silico as disease causing. High-frequency, progressive hearing loss was identified. CONCLUSION: Correlation analysis of pure-tone hearing thresholds revealed progressive hearing loss, especially in the high-frequencies. In the present study, we report the first dominant likely pathogenic variant in MYO3A in a European family and further support MYO3A as an autosomal dominant hearing loss gene.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Umbral Auditivo , Femenino , Genes Dominantes , Pérdida Auditiva Sensorineural/patología , Humanos , Masculino , Mutación Missense , Cadenas Pesadas de Miosina/química , Miosina Tipo III/química , Linaje , Dominios Proteicos
7.
PLoS One ; 15(4): e0230982, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32315303

RESUMEN

Atrial septal defect (ASD) is one of the most common congenital heart defects diagnosed in children. Sarcomeric genes has been attributed to ASD and knockdown of MYH3 functionally homologues gene in chick models indicated abnormal atrial septal development. Here, we report for the first time, a case-control study investigating the role of MYH3 among non-syndromic ASD patients in contributing to septal development. Four amplicons which will amplifies the 40 kb MYH3 were designed and amplified using long range-PCR. The amplicons were then sequenced using indexed paired-end libraries on the MiSeq platform. The STREGA guidelines were applied for planning and reporting. The non-synonymous c. 3574G>A (p.Ala1192Thr) [p = 0.001, OR = 2.30 (1.36-3.87)] located within the tail domain indicated a highly conserved protein region. The mutant model of c. 3574G>A (p.Ala1192Thr) showed high root mean square deviation (RMSD) values compared to the wild model. To our knowledge, this is the first study to provide compelling evidence on the pathogenesis of MYH3 variants towards ASD hence, suggesting the crucial role of non-synonymous variants in the tail domain of MYH3 towards atrial septal development. It is hoped that this gene can be used as panel for diagnosis of ASD in future.


Asunto(s)
Proteínas del Citoesqueleto/genética , Defectos del Tabique Interatrial/genética , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Estudios de Casos y Controles , Niño , Preescolar , Secuencia Conservada , Proteínas del Citoesqueleto/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Genéticos , Modelos Moleculares , Mutación Missense , Cadenas Pesadas de Miosina/química , Miosina Tipo III/química , Polimorfismo de Nucleótido Simple , Adulto Joven
8.
Eur J Med Genet ; 63(5): 103869, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32006683

RESUMEN

We have previously reported on a consanguineous family where 2 siblings, a girl and a boy, presented with tall stature, long and triangular faces, prominent forehead, telecanthus, ptosis, everted lower eyelids, downslanting palpebral fissures, large ears, high arched palate, long arm span, arachnodactyly, advanced bone age, joint laxity, pectus excavatum, inguinal hernia, and myopia, suggestive of a new subtype of connective tissue disorder (Megarbane et al. AJMG, 2012; 158(A)5: 1185-1189). On clinical follow-up, both patients had multiple inguinal, crural, and abdominal herniae, intestinal occlusions, several huge diverticula throughout the gut and the bladder, and rectal prolapse. In addition, the girl had a mild hearing impairment, and the boy a left diaphragmatic hernia. Here we describe the molecular characterization of this disorder using Whole Exome Sequencing, revealing, in both siblings, a novel homozygous missense variant in the EFEMP1 gene, c.163T > C; p.(Cys55Arg) whose homozygous by descent, autosomal recessive transmission was confirmed through segregation analysis by Sanger sequencing. In addition, the girl exhibited a homozygous mutation in the MYO3A gene, c.1370_1371delGA; p.(Arg457Asnfs*25), associated with non-syndromic deafness. The siblings were also found to harbor a homozygous nonsense variant in the VCPKMT gene. We review the literature and discuss our updated clinical and molecular findings that suggest EFEMP1 to be the probable candidate gene implicated in this novel connective tissue disease.


Asunto(s)
Enfermedades del Tejido Conjuntivo/genética , Proteínas de la Matriz Extracelular/genética , Hernia Inguinal/genética , Mutación Missense , Adolescente , Enfermedades del Tejido Conjuntivo/patología , Femenino , Genes Recesivos , Hernia Inguinal/patología , Homocigoto , Humanos , Masculino , Metiltransferasas/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Hermanos , Síndrome , Adulto Joven
9.
Neural Plast ; 2018: 4372913, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30123247

RESUMEN

Myo3a is expressed in cochlear hair cells and retinal cells and is responsible for human recessive hereditary nonsyndromic deafness (DFNB30). To investigate the mechanism of DFNB30-type deafness, we established a mouse model of Myo3a kinase domain Y137C mutation by using CRISPR/Cas9 system. No difference in hearing between 2-month-old Myo3a mutant mice and wild-type mice was observed. The hearing threshold of the ≥6-month-old mutant mice was significantly elevated compared with that of the wild-type mice. We observed degeneration in the inner ear hair cells of 6-month-old Myo3a mutant mice, and the degeneration became more severe at the age of 12 months. We also found structural abnormality in the cochlear hair cell stereocilia. Our results showed that Myo3a is essential for normal hearing by maintaining the intact structure of hair cell stereocilia, and the kinase domain plays a critical role in the normal functions of Myo3a. This mouse line is an excellent model for studying DFNB30-type deafness in humans.


Asunto(s)
Células Ciliadas Auditivas Internas/patología , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/patología , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Estimulación Acústica , Animales , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Técnicas de Sustitución del Gen , Células Ciliadas Auditivas Internas/ultraestructura , Masculino , Mutación
10.
Sci Rep ; 8(1): 8706, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29880844

RESUMEN

Whole-exome sequencing of samples from affected members of two unrelated families with late-onset non-syndromic hearing loss revealed a novel mutation (c.2090 T > G; NM_017433) in MYO3A. The mutation was confirmed in 36 affected individuals, showing autosomal dominant inheritance. The mutation alters a single residue (L697W or p.Leu697Trp) in the motor domain of the stereocilia protein MYO3A, leading to a reduction in ATPase activity, motility, and an increase in actin affinity. MYO3A-L697W showed reduced filopodial actin protrusion initiation in COS7 cells, and a predominant tipward accumulation at filopodia and stereocilia when coexpressed with wild-type MYO3A and espin-1, an actin-regulatory MYO3A cargo. The combined higher actin affinity and duty ratio of the mutant myosin cause increased retention time at stereocilia tips, resulting in the displacement of the wild-type MYO3A protein, which may impact cargo transport, stereocilia length, and mechanotransduction. The dominant negative effect of the altered myosin function explains the dominant inheritance of deafness.


Asunto(s)
Genes Dominantes , Enfermedades Genéticas Congénitas/genética , Pérdida Auditiva/genética , Mutación Missense , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Actinas/genética , Actinas/metabolismo , Adolescente , Adulto , Anciano , Sustitución de Aminoácidos , Animales , Brasil , Células COS , Movimiento Celular/genética , Niño , Chlorocebus aethiops , Femenino , Enfermedades Genéticas Congénitas/metabolismo , Enfermedades Genéticas Congénitas/patología , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Humanos , Masculino , Persona de Mediana Edad , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Seudópodos/genética , Seudópodos/metabolismo , Seudópodos/patología , Estereocilios/genética , Estereocilios/metabolismo , Estereocilios/patología
11.
J Biol Chem ; 291(43): 22781-22792, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27582493

RESUMEN

Class III myosins (MYO3A and MYO3B) are proposed to function as transporters as well as length and ultrastructure regulators within stable actin-based protrusions such as stereocilia and calycal processes. MYO3A differs from MYO3B in that it contains an extended tail domain with an additional actin-binding motif. We examined how the properties of the motor and tail domains of human class III myosins impact their ability to enhance the formation and elongation of actin protrusions. Direct examination of the motor and enzymatic properties of human MYO3A and MYO3B revealed that MYO3A is a 2-fold faster motor with enhanced ATPase activity and actin affinity. A chimera in which the MYO3A tail was fused to the MYO3B motor demonstrated that motor activity correlates with formation and elongation of actin protrusions. We demonstrate that removal of individual exons (30-34) in the MYO3A tail does not prevent filopodia tip localization but abolishes the ability to enhance actin protrusion formation and elongation in COS7 cells. Interestingly, our results demonstrate that MYO3A slows filopodia dynamics and enhances filopodia lifetime in COS7 cells. We also demonstrate that MYO3A is more efficient than MYO3B at increasing formation and elongation of stable microvilli on the surface of cultured epithelial cells. We propose that the unique features of MYO3A, enhanced motor activity, and an extended tail with tail actin-binding motif, allow it to play an important role in stable actin protrusion length and ultrastructure maintenance.


Asunto(s)
Actinas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Seudópodos/metabolismo , Actinas/genética , Animales , Células COS , Chlorocebus aethiops , Humanos , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Seudópodos/genética
12.
Int J Pediatr Otorhinolaryngol ; 84: 43-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27063751

RESUMEN

INTRODUCTION: Hearing loss is a common sensory impairment. Several genetic loci or genes responsible for non-syndrome hearing loss have been identified, including the well-known deafness genes GJB2, MT-RNR1 and SLC26A4. MYO3A belongs to the myosin superfamily. Previously only three mutations in this gene have been found in an Isreali family with DFNB30, in which patients demonstrated progressive hearing loss. METHODS: In this study, we characterized a consanguineous Kazakh family with congenital hearing loss. By targeted sequence capture and next-generation sequencing, we identified a homozygous mutation and did bioinformatics analysis to this mutation. RESULTS: A homozygous mutation, MYO3A:c.1841C>T (p.S614F), was identified to be responsible for the disease. Ser614 is located in the motor domain of MYO3A that is highly conserved among different species. Molecular modeling predicts that the conserved Ser614 may play an important role in maintaining the stability of ß-sheet and the interaction between neighboring ß-strand. CONCLUSIONS: This is the second report on MYO3A mutations in deafness and the first report in China. The finding help facilitate establishing a better relationship between MYO3A mutation and hearing phenotypes.


Asunto(s)
Sordera/genética , Pérdida Auditiva Sensorineural/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Preescolar , China , Consanguinidad , Sordera/congénito , Sordera/etnología , Femenino , Marcadores Genéticos , Pérdida Auditiva Sensorineural/congénito , Pérdida Auditiva Sensorineural/etnología , Homocigoto , Humanos , Kazajstán/etnología , Masculino , Mutación Missense
13.
Nat Commun ; 7: 10833, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26926603

RESUMEN

Hair cells tightly control the dimensions of their stereocilia, which are actin-rich protrusions with graded heights that mediate mechanotransduction in the inner ear. Two members of the myosin-III family, MYO3A and MYO3B, are thought to regulate stereocilia length by transporting cargos that control actin polymerization at stereocilia tips. We show that eliminating espin-1 (ESPN-1), an isoform of ESPN and a myosin-III cargo, dramatically alters the slope of the stereocilia staircase in a subset of hair cells. Furthermore, we show that espin-like (ESPNL), primarily present in developing stereocilia, is also a myosin-III cargo and is essential for normal hearing. ESPN-1 and ESPNL each bind MYO3A and MYO3B, but differentially influence how the two motors function. Consequently, functional properties of different motor-cargo combinations differentially affect molecular transport and the length of actin protrusions. This mechanism is used by hair cells to establish the required range of stereocilia lengths within a single cell.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Estereocilios/fisiología , Animales , Células COS , Chlorocebus aethiops , Oído Interno/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Ratas , Técnicas de Cultivo de Tejidos
14.
Hum Mutat ; 37(5): 481-7, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26841241

RESUMEN

Hereditary hearing loss (HL) is characterized by both allelic and locus genetic heterogeneity. Both recessive and dominant forms of HL may be caused by different mutations in the same deafness gene. In a family with post-lingual progressive non-syndromic deafness, whole-exome sequencing of genomic DNA from five hearing-impaired relatives revealed a single variant, p.Gly488Glu (rs145970949:G>A) in MYO3A, co-segregating with HL as an autosomal dominant trait. This amino acid change, predicted to be pathogenic, alters a highly conserved residue in the motor domain of MYO3A. The mutation severely alters the ATPase activity and motility of the protein in vitro, and the mutant protein fails to accumulate in the filopodia tips in COS7 cells. However, the mutant MYO3A was able to reach the tips of organotypic inner ear culture hair cell stereocilia, raising the possibility of a local effect on positioning of the mechanoelectrical transduction (MET) complex at the stereocilia tips. To address this hypothesis, we investigated the interaction of MYO3A with the cytosolic tail of the integral tip-link protein protocadherin 15 (PCDH15), a core component of MET complex. Interestingly, we uncovered a novel interaction between MYO3A and PCDH15 shedding new light on the function of myosin IIIA at stereocilia tips.


Asunto(s)
Cadherinas/metabolismo , Sordera/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/genética , Miosina Tipo III/metabolismo , Polimorfismo de Nucleótido Simple , Sustitución de Aminoácidos , Animales , Células COS , Proteínas Relacionadas con las Cadherinas , Células Cultivadas , Niño , Preescolar , Chlorocebus aethiops , Sordera/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Linaje
15.
Biochemistry ; 53(49): 7835-45, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25402663

RESUMEN

Motor activity of myosin III is regulated by autophosphorylation. To investigate the role of the kinase activity on the transporter function of myosin IIIA (Myo3A), we identified the phosphorylation sites of kinase domain (KD), which is responsible for the regulation of kinase activity and thus motor function. Using mass spectrometry, we identified six phosphorylation sites in the KD, which are highly conserved among class III myosins and Ste20-related misshapen (Msn) kinases. Two predominant sites, Thr¹84 and Thr¹88, in KD are important for phosphorylation of the KD as well as the motor domain, which regulates the affinity for actin. In the Caco2 cells, the full-length human Myo3A (hMyo3AFull) markedly enlarged the microvilli, although it did not show discrete localization within the microvilli. On the other hand, hMyo3AFull(T184A) and hMyo3AFull(T188A) both showed clear localization at the microvilli tips. Our results suggest that Myo3A induces large actin bundle formation to form microvilli, and phosphorylation of KD at Thr¹84 and Thr¹88 is critical for the kinase activity of Myo3A, and regulation of Myo3A translocation to the tip of microvilli. Retinal extracts potently dephosphorylate both KD and motor domain without IQ motifs (MDIQo), which was inhibited by okadaic acid (OA) with nanomolar range and by tautomycetin (TMC) with micromolar range. The results suggest that Myo3A phosphatase is protein phosphatase type 2A (PP2A). Supporting this result, recombinant PP2Ac potently dephosphorylates both KD and MDIQo. We propose that the phosphorylation-dephosphorylation mechanism plays an essential role in mediating the transport and actin bundle formation and stability functions of hMyo3A.


Asunto(s)
Enterocitos/metabolismo , Microvellosidades/metabolismo , Modelos Moleculares , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Procesamiento Proteico-Postraduccional , Citoesqueleto de Actina/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Células CACO-2 , Dominio Catalítico , Enterocitos/efectos de los fármacos , Enterocitos/ultraestructura , Inhibidores Enzimáticos/farmacología , Furanos/farmacología , Humanos , Lípidos/farmacología , Microvellosidades/efectos de los fármacos , Microvellosidades/ultraestructura , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Cadenas Pesadas de Miosina/antagonistas & inhibidores , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/antagonistas & inhibidores , Miosina Tipo III/química , Miosina Tipo III/genética , Ácido Ocadaico/farmacología , Fosforilación/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Treonina/química
16.
J Biol Chem ; 288(52): 37126-37, 2013 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-24214986

RESUMEN

Class III myosins are unique members of the myosin superfamily in that they contain both a motor and kinase domain. We have found that motor activity is decreased by autophosphorylation, although little is known about the regulation of the kinase domain. We demonstrate by mass spectrometry that Thr-178 and Thr-184 in the kinase domain activation loop and two threonines in the loop 2 region of the motor domain are autophosphorylated (Thr-908 and Thr-919). The kinase activity of MYO3A 2IQ with the phosphomimic (T184E) or phosphoblock (T184A) mutations demonstrates that kinase activity is reduced 30-fold as a result of the T184A mutation, although the Thr-178 site only had a minor impact on kinase activity. Interestingly, the actin-activated ATPase activity of MYO3A 2IQ is slightly reduced as a result of the T178A and T184A mutations suggesting coupling between motor and kinase domains. Full-length GFP-tagged T184A and T184E MYO3A constructs transfected into COS7 cells do not disrupt the ability of MYO3A to localize to filopodia structures. In addition, we demonstrate that T184E MYO3A reduces filopodia elongation in the presence of espin-1, whereas T184A enhances filopodia elongation in a similar fashion to kinase-dead MYO3A. Our results suggest that as MYO3A accumulates at the tips of actin protrusions, autophosphorylation of Thr-184 enhances kinase activity resulting in phosphorylation of the MYO3A motor and reducing motor activity. The differential regulation of the kinase and motor activities allows for MYO3A to precisely self-regulate its concentration in the actin bundle-based structures of cells.


Asunto(s)
Actinas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Seudópodos/enzimología , Actinas/genética , Proteínas Adaptadoras del Transporte Vesicular/genética , Sustitución de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Humanos , Mutación Missense , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/química , Miosina Tipo III/genética , Fosforilación , Estructura Terciaria de Proteína , Seudópodos/genética , Células Sf9 , Spodoptera
17.
J Neurosci ; 32(27): 9205-16, 2012 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-22764229

RESUMEN

Upon illumination several phototransduction proteins translocate between cell body and photosensory compartments. In Drosophila photoreceptors arrestin (Arr2) translocates from cell body to the microvillar rhabdomere down a diffusion gradient created by binding of Arr2 to photo-isomerized metarhodopsin. Translocation is profoundly slowed in mutants of key phototransduction proteins including phospholipase C (PLC) and the Ca(2+)-permeable transient receptor potential channel (TRP), but how the phototransduction cascade accelerates Arr2 translocation is unknown. Using real-time fluorescent imaging of Arr2-green fluorescent protein translocation in dissociated ommatidia, we show that translocation is profoundly slowed in Ca(2+)-free solutions. Conversely, in a blind PLC mutant with ∼100-fold slower translocation, rapid translocation was rescued by the Ca(2+) ionophore, ionomycin. In mutants lacking NINAC (calmodulin [CaM] binding myosin III) in the cell body, translocation remained rapid even in Ca(2+)-free solutions. Immunolabeling revealed that Arr2 in the cell body colocalized with NINAC in the dark. In intact eyes, the impaired translocation found in trp mutants was rescued in ninaC;trp double mutants. Nevertheless, translocation following prolonged dark adaptation was significantly slower in ninaC mutants, than in wild type: a difference that was reflected in the slow decay of the electroretinogram. The results suggest that cytosolic NINAC is a Ca(2+)-dependent binding target for Arr2, which protects Arr2 from immobilization by a second potential sink that sequesters and releases arrestin on a much slower timescale. We propose that rapid Ca(2+)/CaM-dependent release of Arr2 from NINAC upon Ca(2+) influx accounts for the acceleration of translocation by phototransduction.


Asunto(s)
Arrestinas/metabolismo , Calcio/fisiología , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Cadenas Pesadas de Miosina/deficiencia , Miosina Tipo III/fisiología , Células Fotorreceptoras de Invertebrados/metabolismo , Visión Ocular/fisiología , Animales , Animales Modificados Genéticamente , Arrestinas/genética , Señalización del Calcio/genética , Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Proteínas del Ojo/genética , Femenino , Masculino , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Células Fotorreceptoras de Invertebrados/citología , Transporte de Proteínas/fisiología
18.
J Neurochem ; 119(4): 772-84, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21895655

RESUMEN

As class III unconventional myosins are motor proteins with an N-terminal kinase domain, it seems likely they play a role in both signaling and actin based transport. A growing body of evidence indicates that the motor functions of human class IIIA myosin, which has been implicated in progressive hearing loss, are modulated by intermolecular autophosphorylation. However, the phosphorylation sites have not been identified. We studied the kinase activity and phosphorylation sites of mouse class III myosins, mMyo3A and 3B, which are highly similar to their human orthologs. We demonstrate that the kinase domains of mMyo3A and 3B are active kinases, and that they have similar, if not identical, substrate specificities. We show that the kinase domains of these proteins autophosphorylate, and that they can phosphorylate sites within their myosin and tail domains. Using liquid chromatography-mass spectrometry, we identified phosphorylated sites in the kinase, myosin motor and tail domains of both mMyo3A and 3B. Most of the phosphorylated sites we identified and their consensus phosphorylation motifs are highly conserved among vertebrate class III myosins, including human class III myosins. Our findings are a major step toward understanding how the functions of class III myosins are regulated by phosphorylation.


Asunto(s)
Miosina Tipo III/química , Miosina Tipo III/metabolismo , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Aminoácidos , Animales , Humanos , Espectrometría de Masas , Ratones , Miosina Tipo III/clasificación , Miosina Tipo III/genética , Péptidos/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Análisis de Secuencia de Proteína , Especificidad por Sustrato
19.
Mamm Genome ; 22(3-4): 170-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21165622

RESUMEN

The motor protein myosin IIIA is critical for maintenance of normal hearing. Homozygosity and compound heterozygosity for loss-of-function mutations in MYO3A, which encodes myosin IIIA, are responsible for inherited human progressive hearing loss DFNB30. To further evaluate this hearing loss, we constructed a mouse model, Myo3a(KI/KI), that harbors the mutation equivalent to the nonsense allele responsible for the most severe human phenotype. Myo3a(KI/KI) mice were compared to their wild-type littermates. Myosin IIIA, with a unique N-terminal kinase domain and a C-terminal actin-binding domain, localizes to the tips of stereocilia in wild-type mice but is absent in the mutant. The phenotype of the Myo3a(KI/KI) mouse parallels the phenotype of human DFNB30. Hearing loss, as measured by auditory brainstem response, is reduced and progresses significantly with age. Vestibular function is normal. Outer hair cells of Myo3a(KI/KI) mice degenerate with age in a pattern consistent with their progressive hearing loss.


Asunto(s)
Modelos Animales de Enfermedad , Pérdida Auditiva/metabolismo , Ratones , Cadenas Pesadas de Miosina/deficiencia , Miosina Tipo III/deficiencia , Factores de Edad , Animales , Secuencia de Bases , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Pérdida Auditiva/genética , Pérdida Auditiva/fisiopatología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/química , Miosina Tipo III/genética , Estructura Terciaria de Proteína
20.
J Biol Chem ; 285(46): 35770-82, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20826793

RESUMEN

Myosin IIIa (Myo3A) transports cargo to the distal end of actin protrusions and contains a kinase domain that is thought to autoregulate its activity. Because Myo3A tends to cluster at the tips of actin protrusions, we investigated whether intermolecular phosphorylation could regulate Myo3A biochemical activity, cellular localization, and cellular function. Inactivation of Myo3A 2IQ kinase domain with the point mutation K50R did not alter maximal ATPase activity, whereas phosphorylation of Myo3A 2IQ resulted in reduced maximal ATPase activity and actin affinity. The rate and degree of Myo3A 2IQ autophosphorylation was unchanged by the presence of actin but was found to be dependent upon Myo3A 2IQ concentration within the range of 0.1 to 1.2 µm, indicating intermolecular autophosphorylation. In cultured cells, we observed that the filopodial tip localization of Myo3A lacking the kinase domain decreased when co-expressed with kinase-active, full-length Myo3A. The cellular consequence of reduced Myo3A tip localization was decreased filopodial density along the cell periphery, identifying a novel cellular function for Myo3A in mediating the formation and stability of actin-based protrusions. Our results suggest that Myo3A motor activity is regulated through a mechanism involving concentration-dependent autophosphorylation. We suggest that this regulatory mechanism plays an essential role in mediating the transport and actin bundle formation/stability functions of Myo3A.


Asunto(s)
Actinas/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo III/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Ciliadas Vestibulares/metabolismo , Humanos , Microscopía Fluorescente , Mutación , Cadenas Pesadas de Miosina/genética , Miosina Tipo III/genética , Órgano Espiral/metabolismo , Fosforilación , Unión Proteica , Seudópodos/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA