Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunobiology ; 226(6): 152150, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34735924

RESUMO

Macrophages play an important role in maintaining tissue homeostasis, from regulating the inflammatory response to pathogens to resolving inflammation and aiding tissue repair. The surfactant protein A (SP-A) receptor SP-R210 (MYO18A) has been shown to affect basal and inflammatory macrophage states. Specifically, disruption of the longer splice isoform SP-R210L/MYO18Aα renders macrophages hyper-inflammatory, although the mechanism by which this occurs is not well understood. We asked whether disruption of the L isoform led to the hyper-inflammatory state via alteration of global genomic responses. RNA sequencing analysis of L isoform-deficient macrophages (SP-R210L(DN)) revealed basal and influenza-induced upregulation of genes associated with inflammatory pathways, such as TLR, RIG-I, NOD, and cytoplasmic DNA signaling, whereas knockout of both SP-R210 isoforms (L and S) only resulted in increased RIG-I and NOD signaling. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis showed increased genome-wide deposition of the pioneer transcription factor PU.1 in SP-R210L(DN) cells, with increased representation around genes relevant to inflammatory pathways. Additional ChIP-seq analysis of histone H3 methylation marks showed decreases in both repressive H3K9me3 and H3K27me3 marks with a commensurate increase in transcriptionally active (H3K4me3) histone marks in the L isoform deficient macrophages. Influenza A virus (IAV) infection, known to stimulate a wide array of anti-viral responses, caused a differential redistribution of PU.1 binding between proximal promoter and distal sites and decoupling from Toll-like receptor regulated gene promoters in SP-R210L(DN) cells. These finding suggest that the inflammatory differences seen in SP-R210L-deficient macrophages are a result of transcriptional differences that are mediated by epigenetic changes brought about by differential expression of the SP-R210 isoforms. This provides an avenue to explore how the signaling pathways downstream of the receptor and the ligands can modulate the macrophage inflammatory response.


Assuntos
Adaptação Biológica/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Miosinas/genética , Animais , Biomarcadores , Linhagem Celular , Suscetibilidade a Doenças/imunologia , Epigenômica/métodos , Genômica/métodos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunofenotipagem , Camundongos , Miosinas/deficiência , Isoformas de Proteínas , Células RAW 264.7 , Transdução de Sinais
2.
J Immunol ; 206(11): 2521-2526, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34001658

RESUMO

We investigated the function of the newly discovered myosin family protein myosin 18A (Myo18A) in Ab-mediated immunity by generating B cell-conditional Myo18A-deficient mice. Myo18A deficiency led to expansion of bone marrow progenitor B cells and mature B cells in secondary lymphoid organs. Myo18A-deficient mice displayed serum IgM hyperglobulinemia and increased splenic IgM-secreting cells, with older mice switching to IgG1 hyperglobulinemia and autoantibody development. Immunization of Myo18A-deficient mice with inactivated influenza virus led to development of more potent neutralizing Abs against the major Ag hemagglutinin, associated with persistent accumulation of Ag-specific germinal center B cells and more Ag-specific bone marrow plasma cells. In vitro stimulation with TLR7 and BCR ligands revealed a greater ability of Myo18A-deficient B cells to differentiate into Ab-secreting cells, associated with higher AID and Blimp-1 expression. Overall, our study demonstrates that Myo18A is a novel negative regulator of B cell homeostasis, differentiation, and humoral immunity.


Assuntos
Anticorpos Monoclonais/imunologia , Linfócitos B/imunologia , Imunidade Humoral/imunologia , Miosinas/imunologia , Animais , Diferenciação Celular/imunologia , Feminino , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Miosinas/deficiência
3.
BMC Med Genet ; 21(1): 226, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33208113

RESUMO

BACKGROUND: Clinical genetic diagnosis of non-syndromic hearing loss (NSHL) is quite challenging. With regard to its high heterogeneity as well as large size of some genes, it is also really difficult to detect causative mutations using traditional approaches. One of the recent technologies called whole-exome sequencing (WES) has been thus developed in this domain to remove the limitations of conventional methods. METHODS: This study was a report on a research study of two unrelated pedigrees with multiple affected cases of hearing loss (HL). Accordingly, clinical evaluations and genetic analysis were performed in both families. RESULTS: The results of WES data analysis to uncover autosomal recessive non-syndromic hearing loss (ARNSHL) disease-causing variants was reported in the present study. Initial analysis identified two novel variants of MYO15A i.e. c.T6442A:p.W2148R and c.10504dupT:p.C3502Lfs*15 correspondingly which were later confirmed by Sanger validations and segregation analyses. According to online prediction tools, both identified variants seemed to have damaging effects. CONCLUSION: In this study, whole exome sequencing were used as a first approach strategy to identify the two novel variants in MYO15A in two Iranian families with ARNSHL.


Assuntos
Surdez/genética , Perda Auditiva Neurossensorial/genética , Mutação , Miosinas/genética , Adolescente , Adulto , Sequência de Bases , Consanguinidade , Surdez/diagnóstico , Surdez/patologia , Feminino , Expressão Gênica , Genes Recessivos , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/patologia , Humanos , Irã (Geográfico) , Masculino , Miosinas/deficiência , Linhagem , Sequenciamento do Exoma
4.
J Bone Miner Res ; 34(5): 939-954, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30645777

RESUMO

Normal bone mass is maintained by balanced bone formation and resorption. Myosin X (Myo10), an unconventional "myosin tail homology 4-band 4.1, ezrin, radixin, moesin" (MyTH4-FERM) domain containing myosin, is implicated in regulating osteoclast (OC) adhesion, podosome positioning, and differentiation in vitro. However, evidence is lacking for Myo10 in vivo function. Here we show that mice with Myo10 loss of function, Myo10m/m , exhibit osteoporotic deficits, which are likely due to the increased OC genesis and bone resorption because bone formation is unchanged. Similar deficits are detected in OC-selective Myo10 conditional knockout (cko) mice, indicating a cell autonomous function of Myo10. Further mechanistic studies suggest that Unc-5 Netrin receptor B (Unc5b) protein levels, in particular its cell surface level, are higher in the mutant OCs, but lower in RAW264.7 cells or HEK293 cells expressing Myo10. Suppressing Unc5b expression in bone marrow macrophages (BMMs) from Myo10m/m mice by infection with lentivirus of Unc5b shRNA markedly impaired RANKL-induced OC genesis. Netrin-1, a ligand of Unc5b, increased RANKL-induced OC formation in BMMs from both wild-type and Myo10m/m mice. Taken together, these results suggest that Myo10 plays a negative role in OC formation, likely by inhibiting Unc5b cell-surface targeting, and suppressing Netrin-1 promoted OC genesis. © 2019 American Society for Bone and Mineral Research.


Assuntos
Miosinas/metabolismo , Receptores de Netrina/metabolismo , Osteoclastos/metabolismo , Osteoporose/metabolismo , Acebutolol , Animais , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Miosinas/deficiência , Receptores de Netrina/genética , Netrina-1/genética , Netrina-1/metabolismo , Osteoclastos/patologia , Osteoporose/genética , Osteoporose/patologia , Ligante RANK/genética , Ligante RANK/metabolismo , Células RAW 264.7
5.
J Exp Med ; 215(7): 1869-1890, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29875261

RESUMO

T cells are actively scanning pMHC-presenting cells in lymphoid organs and nonlymphoid tissues (NLTs) with divergent topologies and confinement. How the T cell actomyosin cytoskeleton facilitates this task in distinct environments is incompletely understood. Here, we show that lack of Myosin IXb (Myo9b), a negative regulator of the small GTPase Rho, led to increased Rho-GTP levels and cell surface stiffness in primary T cells. Nonetheless, intravital imaging revealed robust motility of Myo9b-/- CD8+ T cells in lymphoid tissue and similar expansion and differentiation during immune responses. In contrast, accumulation of Myo9b-/- CD8+ T cells in NLTs was strongly impaired. Specifically, Myo9b was required for T cell crossing of basement membranes, such as those which are present between dermis and epidermis. As consequence, Myo9b-/- CD8+ T cells showed impaired control of skin infections. In sum, we show that Myo9b is critical for the CD8+ T cell adaptation from lymphoid to NLT surveillance and the establishment of protective tissue-resident T cell populations.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Miosinas/metabolismo , Animais , Linfócitos T CD8-Positivos/citologia , Movimento Celular , Polaridade Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Epiderme/patologia , Epiderme/virologia , Matriz Extracelular/metabolismo , Imunidade , Ativação Linfocitária/imunologia , Tecido Linfoide/metabolismo , Camundongos Endogâmicos C57BL , Miosinas/deficiência , Receptores de Retorno de Linfócitos/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
6.
J Mech Behav Biomed Mater ; 84: 99-107, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29754047

RESUMO

The objective of this study was to examine relationships among a variety of bone characteristics, including volumetric, mineral density, geometric, dynamic mechanical analysis, and static fracture mechanical properties. As MYO9B is an unconventional myosin in bone cells responsible for normal skeletal growth, bone characteristics of wild-type (WT), heterozygous (HET), and MYO9B knockout (KO) mice groups were compared as an animal model to express different bone quantity and quality. Forty-five sex-matched 12-week-old mice were used in this study. After euthanization, femurs were isolated and scanned using microcomputed tomography (micro-CT) to assess bone volumetric, tissue mineral density (TMD), and geometric parameters. Then, a non-destructive dynamic mechanical analysis (DMA) was performed by applying oscillatory bending displacement on the femur. Finally, the same femur was subject to static fracture testing. KO group had significantly lower length, bone mineral density (BMD), bone mass and volume, dynamic and static stiffness, and strength than WT and HET groups (p < 0.019). On the other hand, TMD parameters of KO group were comparable with those of WT group. HET group showed volumetric, geometric, and mechanical properties similar to WT group, but had lower TMD (p < 0.014). Non-destructive micro-CT and DMA parameters had significant positive correlations with strength (p < 0.015) without combined effect of groups and sex on the correlations (p > 0.077). This comprehensive characterization provides a better understanding of interactive behavior between the tissue- and organ-level of the same femur. The current findings elucidate that MYO9B is responsible for controlling bone volume to determine the growth rate and fracture risk of bone.


Assuntos
Fêmur/metabolismo , Técnicas de Inativação de Genes , Fenômenos Mecânicos , Miosinas/deficiência , Miosinas/genética , Animais , Fenômenos Biomecânicos , Densidade Óssea/genética , Fêmur/fisiologia , Camundongos
7.
Hum Mol Genet ; 27(8): 1434-1446, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29462312

RESUMO

Congenital myasthenic syndromes (CMS) are a group of rare, inherited disorders characterized by compromised function of the neuromuscular junction, manifesting with fatigable muscle weakness. Mutations in MYO9A were previously identified as causative for CMS but the precise pathomechanism remained to be characterized. On the basis of the role of MYO9A as an actin-based molecular motor and as a negative regulator of RhoA, we hypothesized that loss of MYO9A may affect the neuronal cytoskeleton, leading to impaired intracellular transport. To investigate this, we used MYO9A-depleted NSC-34 cells (mouse motor neuron-derived cells), revealing altered expression of a number of cytoskeletal proteins important for neuron structure and intracellular transport. On the basis of these findings, the effect on protein transport was determined using a vesicular recycling assay which revealed impaired recycling of a neuronal growth factor receptor. In addition, an unbiased approach utilizing proteomic profiling of the secretome revealed a key role for defective intracellular transport affecting proper protein secretion in the pathophysiology of MYO9A-related CMS. This also led to the identification of agrin as being affected by the defective transport. Zebrafish with reduced MYO9A orthologue expression were treated with an artificial agrin compound, ameliorating defects in neurite extension and improving motility. In summary, loss of MYO9A affects the neuronal cytoskeleton and leads to impaired transport of proteins, including agrin, which may provide a new and unexpected treatment option.


Assuntos
Agrina/metabolismo , Neurônios Motores/metabolismo , Debilidade Muscular/genética , Síndromes Miastênicas Congênitas/genética , Miosinas/genética , Fator de Crescimento Neural/genética , Junção Neuromuscular/metabolismo , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestrutura , Actinas/genética , Actinas/metabolismo , Agrina/genética , Amidas , Animais , Movimento Celular , Modelos Animais de Doenças , Embrião não Mamífero , Inibidores Enzimáticos , Regulação da Expressão Gênica , Humanos , Filamentos Intermediários/genética , Filamentos Intermediários/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Neurônios Motores/ultraestrutura , Debilidade Muscular/metabolismo , Debilidade Muscular/patologia , Síndromes Miastênicas Congênitas/metabolismo , Síndromes Miastênicas Congênitas/patologia , Miosinas/deficiência , Fator de Crescimento Neural/metabolismo , Junção Neuromuscular/ultraestrutura , Transporte Proteico , Piridinas , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Peixe-Zebra , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP
8.
EMBO Rep ; 18(9): 1521-1535, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28710093

RESUMO

Primary cilia are sensory, antennae-like organelles present on the surface of many cell types. They have been involved in a variety of diseases collectively termed ciliopathies. As cilia are essential regulators of cell signaling, the composition of the ciliary membrane needs to be strictly regulated. To understand regulatory processes at the ciliary membrane, we report the targeting of a genetically engineered enzyme specifically to the ciliary membrane to allow biotinylation and identification of the membrane-associated proteome. Bioinformatic analysis of the comprehensive dataset reveals high-stoichiometric presence of actin-binding proteins inside the cilium. Immunofluorescence stainings and complementary interaction proteomic analyses confirm these findings. Depolymerization of branched F-actin causes further enrichment of the actin-binding and actin-related proteins in cilia, including Myosin 5a (Myo5a). Interestingly, Myo5a knockout decreases ciliation while enhanced levels of Myo5a are observed in cilia upon induction of ciliary disassembly. In summary, we present a novel approach to investigate dynamics of the ciliary membrane proteome in mammalian cells and identify actin-binding proteins as mechanosensitive components of cilia that might have important functions in cilia membrane dynamics.


Assuntos
Actinas/metabolismo , Cílios/metabolismo , Proteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteoma/metabolismo , Actinas/química , Animais , Biologia Computacional , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Membranas/metabolismo , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Miosinas/deficiência , Miosinas/genética , Miosinas/metabolismo , Proteômica , Transdução de Sinais
9.
J Bone Miner Res ; 32(10): 2103-2115, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28585695

RESUMO

The Ras homolog A (RhoA) subfamily of Rho guanosine triphosphatases (GTPases) regulates actin-based cellular functions in bone such as differentiation, migration, and mechanotransduction. Polymorphisms or genetic ablation of RHOA and some of its regulatory guanine exchange factors (GEFs) have been linked to poor bone health in humans and mice, but the effects of RhoA-specific GTPase-activating proteins (GAPs) on bone quality have not yet been identified. Therefore, we examined the consequences of RhoGAP Myo9b gene knockout on bone growth, phenotype, and cellular activity. Male and female mice lacking both alleles demonstrated growth retardation and decreased bone formation rates during early puberty. These mice had smaller, weaker bones by 4 weeks of age, but only female KOs had altered cellular numbers, with fewer osteoblasts and more osteoclasts. By 12 weeks of age, bone quality in KOs worsened. In contrast, 4-week-old heterozygotes demonstrated bone defects that resolved by 12 weeks of age. Throughout, Myo9b ablation affected females more than males. Osteoclast activity appeared unaffected. In primary osteogenic cells, Myo9b was distributed in stress fibers and focal adhesions, and its absence resulted in poor spreading and eventual detachment from culture dishes. Similarly, MC3T3-E1 preosteoblasts with transiently suppressed Myo9b levels spread poorly and contained decreased numbers of focal adhesions. These cells also demonstrated reduced ability to undergo IGF-1-induced spreading or chemotaxis toward IGF-1, though responses to PDGF and BMP-2 were unaffected. IGF-1 receptor (IGF1R) activation was normal in cells with diminished Myo9b levels, but the activated receptor was redistributed from stress fibers and focal adhesions into nuclei, potentially affecting receptor accessibility and gene expression. These results demonstrate that Myo9b regulates a subset of RhoA-activated processes necessary for IGF-1 responsiveness in osteogenic cells, and is critical for normal bone formation in growing mice. © 2017 American Society for Bone and Mineral Research.


Assuntos
Desenvolvimento Ósseo , Fator de Crescimento Insulin-Like I/farmacologia , Miosinas/metabolismo , Osteoblastos/metabolismo , Animais , Fenômenos Biomecânicos , Desenvolvimento Ósseo/efeitos dos fármacos , Osso Esponjoso/metabolismo , Osso Esponjoso/patologia , Osso Esponjoso/fisiopatologia , Adesão Celular , Linhagem Celular , Quimiotaxia , Fêmur/metabolismo , Fêmur/patologia , Fêmur/fisiopatologia , Técnicas de Silenciamento de Genes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosinas/deficiência , Osteoblastos/efeitos dos fármacos , Ratos , Maturidade Sexual
10.
Circ Res ; 120(5): 799-815, 2017 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-28115390

RESUMO

RATIONALE: Abdominal aortic aneurysms (AAAs) are characterized by pathological remodeling of the aortic wall. Although both increased Krüppel-like factor 5 (KLF5) expression and macrophage infiltration have been implicated in vascular remodeling, the role of KLF5 in macrophage infiltration and AAA formation remains unclear. OBJECTIVE: To determine the role of KLF5 in AAA formation and macrophage infiltration into AAAs. METHODS AND RESULTS: KLF5 expression was significantly increased in human AAA tissues and in 2 mouse models of experimental AAA. Moreover, in myeloid-specific Klf5 knockout mice (myeKlf5-/- mice), macrophage infiltration, medial smooth muscle cell loss, elastin degradation, and AAA formation were markedly decreased. In cell migration and time-lapse imaging analyses, the migration of murine myeKlf5-/- macrophages was impaired, and in luciferase reporter assays, KLF5 activated Myo9b (myosin IXB) transcription by direct binding to the Myo9b promoter. In subsequent coimmunostaining studies, Myo9b was colocalized with filamentous actin, cortactin, vinculin, and Tks5 in the podosomes of phorbol 12,13-dibutyrate-treated macrophages, indicating that Myo9b participates in podosome formation. Gain- and loss-of-function experiments showed that KLF5 promoted podosome formation in macrophages by upregulating Myo9b expression. Furthermore, RhoA-GTP levels increased after KLF5 knockdown in macrophages, suggesting that KLF5 lies upstream of RhoA signaling. Finally, Myo9b expression was increased in human AAA tissues, located in macrophages, and positively correlated with AAA size. CONCLUSIONS: These data are the first to indicate that KLF5-dependent regulation of Myo9b/RhoA is required for podosome formation and macrophage migration during AAA formation, warranting consideration of the KLF5-Myo9b-RhoA pathway as a therapeutic target for AAA treatment.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/prevenção & controle , Fatores de Transcrição Kruppel-Like/biossíntese , Macrófagos/metabolismo , Miosinas/biossíntese , Podossomos/metabolismo , Proteína rhoA de Ligação ao GTP/biossíntese , Animais , Linhagem Celular , Células HEK293 , Humanos , Fatores de Transcrição Kruppel-Like/deficiência , Masculino , Camundongos , Camundongos Knockout , Miosinas/deficiência , Transdução de Sinais/fisiologia , Proteína rhoA de Ligação ao GTP/deficiência
11.
Hear Res ; 343: 14-33, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27473502

RESUMO

Studies of congenital and early-onset deafness have demonstrated that an absence of peripheral sound-evoked activity in the auditory nerve causes pathological changes in central auditory structures. The aim of this study was to establish whether progressive acquired hearing loss could lead to similar brain changes that would degrade the precision of signal transmission. We used complementary physiologic hearing tests and microscopic techniques to study the combined effect of both magnitude and duration of hearing loss on one of the first auditory synapses in the brain, the endbulb of Held (EB), along with its bushy cell (BC) target in the anteroventral cochlear nucleus. We compared two hearing mouse strains (CBA/Ca and heterozygous shaker-2+/-) against a model of early-onset progressive hearing loss (DBA/2) and a model of congenital deafness (homozygous shaker-2-/-), examining each strain at 1, 3, and 6 months of age. Furthermore, we employed a frequency model of the mouse cochlear nucleus to constrain our analyses to regions most likely to exhibit graded changes in hearing function with time. No significant differences in the gross morphology of EB or BC structure were observed in 1-month-old animals, indicating uninterrupted development. However, in animals with hearing loss, both EBs and BCs exhibited a graded reduction in size that paralleled the hearing loss, with the most severe pathology seen in deaf 6-month-old shaker-2-/- mice. Ultrastructural pathologies associated with hearing loss were less dramatic: minor changes were observed in terminal size but mitochondrial fraction and postsynaptic densities remained relatively stable. These results indicate that acquired progressive hearing loss can have consequences on auditory brain structure, with prolonged loss leading to greater pathologies. Our findings suggest a role for early intervention with assistive devices in order to mitigate long-term pathology and loss of function.


Assuntos
Nervo Coclear/ultraestrutura , Núcleo Coclear/ultraestrutura , Perda Auditiva/patologia , Audição , Sinapses/ultraestrutura , Estimulação Acústica , Fatores Etários , Animais , Limiar Auditivo , Comportamento Animal , Nervo Coclear/fisiopatologia , Núcleo Coclear/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Predisposição Genética para Doença , Audição/genética , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Perda Auditiva/psicologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Miosinas/deficiência , Miosinas/genética , Fenótipo , Índice de Gravidade de Doença , Fatores de Tempo
12.
Hear Res ; 343: 34-49, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27421755

RESUMO

Auditory efferent neurons reside in the brain and innervate the sensory hair cells of the cochlea to modulate incoming acoustic signals. Two groups of efferents have been described in mouse and this report will focus on the medial olivocochlear (MOC) system. Electrophysiological data suggest the MOC efferents function in selective listening by differentially attenuating auditory nerve fiber activity in quiet and noisy conditions. Because speech understanding in noise is impaired in age-related hearing loss, we asked whether pathologic changes in input to MOC neurons from higher centers could be involved. The present study investigated the anatomical nature of descending projections from the inferior colliculus (IC) to MOCs in 3-month old mice with normal hearing, and in 6-month old mice with normal hearing (CBA/CaH), early onset progressive hearing loss (DBA/2), and congenital deafness (homozygous Shaker-2). Anterograde tracers were injected into the IC and retrograde tracers into the cochlea. Electron microscopic analysis of double-labelled tissue confirmed direct synaptic contact from the IC onto MOCs in all cohorts. These labelled terminals are indicative of excitatory neurotransmission because they contain round synaptic vesicles, exhibit asymmetric membrane specializations, and are co-labelled with antibodies against VGlut2, a glutamate transporter. 3D reconstructions of the terminal fields indicate that in normal hearing mice, descending projections from the IC are arranged tonotopically with low frequencies projecting laterally and progressively higher frequencies projecting more medially. Along the mediolateral axis, the projections of DBA/2 mice with acquired high frequency hearing loss were shifted medially towards expected higher frequency projecting regions. Shaker-2 mice with congenital deafness had a much broader spatial projection, revealing abnormalities in the topography of connections. These data suggest that loss in precision of IC directed MOC activation could contribute to impaired signal detection in noise.


Assuntos
Cóclea/inervação , Surdez/fisiopatologia , Audição , Colículos Inferiores/fisiopatologia , Núcleo Olivar/fisiopatologia , Estimulação Acústica , Animais , Vias Auditivas/fisiopatologia , Percepção Auditiva , Comportamento Animal , Surdez/metabolismo , Surdez/patologia , Surdez/psicologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Predisposição Genética para Doença , Audição/genética , Colículos Inferiores/metabolismo , Colículos Inferiores/ultraestrutura , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Miosinas/deficiência , Miosinas/genética , Técnicas de Rastreamento Neuroanatômico , Núcleo Olivar/metabolismo , Núcleo Olivar/ultraestrutura , Fenótipo , Detecção de Sinal Psicológico , Sinapses/ultraestrutura , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
13.
Infect Immun ; 84(5): 1556-64, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26953327

RESUMO

Myosins are a family of actin-based motor proteins found in many organisms and are categorized into classes based on their structures. Class II and V myosins are known to be important for critical cellular processes, including cytokinesis, endocytosis, exocytosis, and organelle trafficking, in the model fungi Saccharomyces cerevisiae and Aspergillus nidulans However, the roles of myosins in the growth and virulence of the pathogen Aspergillus fumigatus are unknown. We constructed single- and double-deletion strains of the class II and class V myosins in A. fumigatus and found that while the class II myosin (myoB) is dispensable for growth, the class V myosin (myoE) is required for proper hyphal extension; deletion of myoE resulted in hyperbranching and loss of hyphal polarity. Both myoB and myoE are necessary for proper septation, conidiation, and conidial germination, but only myoB is required for conidial viability. Infection with the ΔmyoE strain in the invertebrate Galleria mellonella model and also in a persistently immunosuppressed murine model of invasive aspergillosis resulted in hypovirulence, while analysis of bronchoalveolar lavage fluid revealed that tumor necrosis factor alpha (TNF-α) release and cellular infiltration were similar compared to those of the wild-type strain. The ΔmyoE strain showed fungal growth in the murine lung, while the ΔmyoB strain exhibited little fungal burden, most likely due to the reduced conidial viability. These results show, for the first time, the important role these cytoskeletal components play in the growth of and disease caused by a known pathogen, prompting future studies to understand their regulation and potential targeting for novel antifungal therapies.


Assuntos
Aspergillus fumigatus/crescimento & desenvolvimento , Aspergillus fumigatus/patogenicidade , Proteínas Fúngicas/metabolismo , Hifas/crescimento & desenvolvimento , Miosinas/metabolismo , Animais , Aspergilose/microbiologia , Aspergilose/patologia , Aspergillus fumigatus/genética , Contagem de Colônia Microbiana , Proteínas Fúngicas/genética , Técnicas de Inativação de Genes , Pulmão/microbiologia , Masculino , Camundongos , Viabilidade Microbiana , Miosinas/deficiência , Esporos Fúngicos/crescimento & desenvolvimento , Virulência
14.
J Struct Biol ; 194(2): 139-46, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26806019

RESUMO

All inner ear organs possess extracellular matrix appendices over the sensory epithelia that are crucial for their proper function. The tectorial membrane (TM) is a gelatinous acellular membrane located above the hearing sensory epithelium and is composed mostly of type II collagen, and α and ß tectorins. TM molecules self-assemble in the endolymph fluid environment, interacting medially with the spiral limbus and distally with the outer hair cell stereocilia. Here, we used immunogold labeling in freeze-substituted mouse cochleae to assess the fine localization of both tectorins in distinct TM regions. We observed that the TM adheres to the spiral limbus through a dense thin matrix enriched in α- and ß-tectorin, both likely bound to the membranes of interdental cells. Freeze-etching images revealed that type II collagen fibrils were crosslinked by short thin filaments (4±1.5nm, width), resembling another collagen type protein, or chains of globular elements (15±3.2nm, diameter). Gold-particles for both tectorins also localized adjacent to the type II collagen fibrils, suggesting that these globules might be composed essentially of α- and ß-tectorins. Finally, the presence of gold-particles at the TM lower side suggests that the outer hair cell stereocilia membrane has a molecular partner to tectorins, probably stereocilin, allowing the physical connection between the TM and the organ of Corti.


Assuntos
Colágeno Tipo II/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteínas de Membrana/metabolismo , Órgão Espiral/metabolismo , Membrana Tectorial/metabolismo , Animais , Colágeno Tipo II/genética , Colágeno Tipo II/ultraestrutura , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/ultraestrutura , Técnica de Congelamento e Réplica , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/ultraestrutura , Expressão Gênica , Cobaias , Imuno-Histoquímica , Proteínas de Membrana/genética , Proteínas de Membrana/ultraestrutura , Camundongos , Microscopia Eletrônica de Transmissão , Miosinas/deficiência , Miosinas/genética , Órgão Espiral/ultraestrutura , Ligação Proteica , Ratos , Membrana Tectorial/ultraestrutura
15.
Brain Res ; 1620: 139-52, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25988834

RESUMO

Studies of human brain malformations, such as lissencephaly and double cortex, have revealed the importance of neuronal migration during cortical development. Afadin, a membrane scaffolding protein, regulates the formation of adherens junctions (AJs) and cell migration to form and maintain tissue structures. Here, we report that mice with dorsal telencephalon-specific ablation of afadin gene exhibited defects similar to human double cortex, in which the heterotopic cortex was located underneath the normotopic cortex. The normotopic cortex of the mutant mice was arranged in the pattern similar to the cortex of the control mice, while the heterotopic cortex was disorganized. As seen in human patients, double cortex in the mutant mice was formed by impaired neuronal migration during cortical development. Genetic ablation of afadin in the embryonic cerebral cortex disrupted AJs of radial glial cells, likely resulting in the retraction of the apical endfeet from the ventricular surface and the dispersion of radial glial cells from the ventricular zone to the subventricular and intermediate zones. These results indicate that afadin is required for the maintenance of AJs of radial glial cells and that the disruption of AJs might cause an abnormal radial scaffold for neuronal migration. In contrast, the proliferation or differentiation of radial glial cells was not significantly affected. Taken together, these findings indicate that afadin is required for the maintenance of the radial glial scaffold for neuronal migration and that the genetic ablation of afadin leads to the formation of double cortex.


Assuntos
Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/fisiopatologia , Cinesinas/deficiência , Miosinas/deficiência , Neuroglia/fisiologia , Neurônios/fisiologia , Animais , Animais Recém-Nascidos , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/patologia , Modelos Animais de Doenças , Imunofluorescência , Cinesinas/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Miosinas/genética , Neuroglia/patologia , Neurônios/patologia
16.
Eur J Immunol ; 45(1): 225-37, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25263281

RESUMO

TLR-mediated recognition of microbial danger induces substantial changes in macrophage migration, adherence, and phagocytosis. Recently, we described the LPS-regulated phosphorylation of many cytoskeleton-associated proteins by phosphoproteomics. The functional role of these cytoskeletal and motor proteins in innate immune cell responses is largely unexplored. Here, we first identified both long-tailed class I myosins Myo1e and Myo1f as important contributors to LPS-triggered macrophage spreading. Mouse bone marrow-derived macrophages and DCs deficient in Myo1e selectively secreted increased amounts of the chemokine CCL2. In addition, the cell surface expression of MHC class II (MHC-II) on both cell types was reduced in the absence of Myo1e. However, transcriptional changes in CCL2 and MHC-II were not observed in the absence of Myo1e, indicating that Myo1e regulates specific intracellular transport processes. The capacity of macrophages and DCs lacking Myo1e to stimulate antigen-specific CD4(+) T-cell proliferation was impaired, consistent with the reduced MHC-II surface protein levels. Surprisingly, in Myo1e-deficient DCs, the proteolytic cleavage of endocytosed antigen was also increased. Together, our results provide evidence for a non-redundant function of the motor protein Myo1e in the regulation of TLR4-controlled, cytoskeleton-associated functional properties of macrophages and DCs, and in induction of a full MHC-II-restricted adaptive immune response.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Macrófagos/imunologia , Miosinas/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Apresentação de Antígeno/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Proliferação de Células , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Citoesqueleto/química , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/imunologia , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina Tipo I/genética , Miosina Tipo I/imunologia , Miosinas/deficiência , Miosinas/genética , Cultura Primária de Células , Transdução de Sinais , Receptor 4 Toll-Like/genética
17.
J Genet Genomics ; 41(8): 417-28, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25160974

RESUMO

Myosin XVIIIA, or MYO18A, is a unique PDZ domain-containing unconventional myosin and is evolutionarily conserved from Drosophila to vertebrates. Although there is evidence indicating its expression in the somites, whether it regulates muscle function remains unclear. We show that the two zebrafish myo18a genes (myo18aa and myo18ab) are predominantly expressed at somite borders during early developmental stages. Knockdown of these genes or overexpression of the MYO18A PDZ domain disrupts myofiber integrity, induces myofiber lesions, and compromises the localization of dystrophin, α-dystroglycan (α-DG) and laminin at the myotome boundaries. Cell transplantation experiments indicate that myo18a morphant myoblasts fail to form elongated myofibers in the myotomes of wild-type embryos, which can be rescued by the full-length MYO18A protein. These results suggest that MYO18A likely functions in the adhesion process that maintains the stable attachment of myofibers to ECM (extracellular matrix) and muscle integrity during early development.


Assuntos
Embrião não Mamífero/metabolismo , Músculos/embriologia , Músculos/metabolismo , Miosinas/química , Miosinas/metabolismo , Domínios PDZ , Peixe-Zebra/embriologia , Animais , Adesão Celular , Distroglicanas/metabolismo , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/metabolismo , Embrião não Mamífero/citologia , Técnicas de Silenciamento de Genes , Laminas/metabolismo , Músculos/citologia , Mioblastos/citologia , Mioblastos/metabolismo , Miosinas/deficiência , Miosinas/genética , Transporte Proteico , Somitos/citologia , Somitos/metabolismo
18.
Circ Res ; 114(6): 982-92, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24508725

RESUMO

RATIONALE: Kv1.5 (KCNA5) mediates the ultra-rapid delayed rectifier current that controls atrial action potential duration. Given its atrial-specific expression and alterations in human atrial fibrillation, Kv1.5 has emerged as a promising target for the treatment of atrial fibrillation. A necessary step in the development of novel agents that selectively modulate trafficking pathways is the identification of the cellular machinery controlling Kv1.5 surface density, of which little is yet known. OBJECTIVE: To investigate the role of the unconventional myosin-V (MYO5A and MYO5B) motors in determining the cell surface density of Kv1.5. METHODS AND RESULTS: Western blot analysis showed MYO5A and MYO5B expression in the heart, whereas disruption of endogenous motors selectively reduced IKur current in adult rat cardiomyocytes. Dominant negative constructs and short hairpin RNA silencing demonstrated a role for MYO5A and MYO5B in the surface trafficking of Kv1.5 and connexin-43 but not potassium voltage-gated channel, subfamily H (eag-related), member 2 (KCNH2). Live-cell imaging of Kv1.5-GFP and retrospective labeling of phalloidin demonstrated motility of Kv1.5 vesicles on actin tracts. MYO5A participated in anterograde trafficking, whereas MYO5B regulated postendocytic recycling. Overexpression of mutant motors revealed a selective role for Rab11 in coupling MYO5B to Kv1.5 recycling. CONCLUSIONS: MYO5A and MYO5B control functionally distinct steps in the surface trafficking of Kv1.5. These isoform-specific trafficking pathways determine Kv1.5-encoded IKur in myocytes to regulate repolarizing current and, consequently, cardiac excitability. Therapeutic strategies that manipulate Kv1.5 selective trafficking pathways may prove useful in the treatment of arrhythmias.


Assuntos
Membrana Celular/metabolismo , Canal de Potássio Kv1.5/metabolismo , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/fisiologia , Miosina Tipo V/fisiologia , Miosinas/fisiologia , Transporte Proteico/fisiologia , Citoesqueleto de Actina/fisiologia , Animais , Arritmias Cardíacas/fisiopatologia , Linhagem Celular , Conexina 43/análise , Canal de Potássio ERG1 , Endocitose , Canais de Potássio Éter-A-Go-Go/análise , Junções Comunicantes , Genes Reporter , Sistema de Condução Cardíaco/fisiopatologia , Transporte de Íons , Canal de Potássio Kv1.5/genética , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Cardiovasculares , Cadeias Pesadas de Miosina/deficiência , Cadeias Pesadas de Miosina/genética , Miosina Tipo V/deficiência , Miosina Tipo V/genética , Miosinas/deficiência , Miosinas/genética , Potássio/metabolismo , Isoformas de Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Proteínas rab de Ligação ao GTP/fisiologia
19.
PLoS One ; 8(8): e72027, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991031

RESUMO

Mutations in MYO7A cause autosomal recessive Usher syndrome type IB (USH1B), one of the most frequent conditions that combine severe congenital hearing impairment and retinitis pigmentosa. A promising therapeutic strategy for retinitis pigmentosa is gene therapy, however its pre-clinical development is limited by the mild retinal phenotype of the shaker1 (sh1(-/-)) murine model of USH1B which lacks both retinal functional abnormalities and degeneration. Here we report a significant, early-onset delay of sh1(-/-) photoreceptor ability to recover from light desensitization as well as a progressive reduction of both b-wave electroretinogram amplitude and light sensitivity, in the absence of significant loss of photoreceptors up to 12 months of age. We additionally show that subretinal delivery to the sh1(-/-) retina of AAV vectors encoding the large MYO7A protein results in significant improvement of sh1(-/-) photoreceptor and retinal pigment epithelium ultrastructural anomalies which is associated with improvement of recovery from light desensitization. These findings provide new tools to evaluate the efficacy of experimental therapies for USH1B. In addition, although AAV vectors expressing large genes might have limited clinical applications due to their genome heterogeneity, our data show that AAV-mediated MYO7A gene transfer to the sh1(-/-) retina is effective.


Assuntos
Terapia Genética/métodos , Miosinas/deficiência , Retina/fisiopatologia , Degeneração Retiniana/terapia , Adulto , Animais , Western Blotting , Dependovirus/genética , Modelos Animais de Doenças , Eletrorretinografia , Olho/metabolismo , Olho/fisiopatologia , Olho/ultraestrutura , Feminino , Vetores Genéticos/genética , Células HEK293 , Humanos , Masculino , Melanossomas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Microscopia Eletrônica , Miosina VIIa , Miosinas/genética , Miosinas/metabolismo , Retina/metabolismo , Retina/ultraestrutura , Degeneração Retiniana/genética , Degeneração Retiniana/fisiopatologia , Síndromes de Usher/genética , Síndromes de Usher/fisiopatologia , Síndromes de Usher/terapia
20.
J Cell Biol ; 199(2): 381-99, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-23045546

RESUMO

The mechanisms underlying retinal dystrophy in Usher syndrome type I (USH1) remain unknown because mutant mice lacking any of the USH1 proteins-myosin VIIa, harmonin, cadherin-23, protocadherin-15, sans-do not display retinal degeneration. We found here that, in macaque photoreceptor cells, all USH1 proteins colocalized at membrane interfaces (i) between the inner and outer segments in rods and (ii) between the microvillus-like calyceal processes and the outer segment basolateral region in rods and cones. This pattern, conserved in humans and frogs, was mediated by the formation of an USH1 protein network, which was associated with the calyceal processes from the early embryonic stages of outer segment growth onwards. By contrast, mouse photoreceptors lacked calyceal processes and had no USH1 proteins at the inner-outer segment interface. We suggest that USH1 proteins form an adhesion belt around the basolateral region of the photoreceptor outer segment in humans, and that defects in this structure cause the retinal degeneration in USH1 patients.


Assuntos
Junções Intercelulares/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/ultraestrutura , Síndromes de Usher/metabolismo , Animais , Anuros , Proteínas Relacionadas a Caderinas , Caderinas/deficiência , Caderinas/genética , Caderinas/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Humanos , Junções Intercelulares/ultraestrutura , Macaca fascicularis , Camundongos , Miosina VIIa , Miosinas/deficiência , Miosinas/genética , Miosinas/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Precursores de Proteínas/deficiência , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Retina/metabolismo , Retina/ultraestrutura , Distrofias Retinianas/patologia , Suínos , Síndromes de Usher/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...