Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 42(17): e114415, 2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37427462

RESUMO

Cell fragmentation is commonly observed in human preimplantation embryos and is associated with poor prognosis during assisted reproductive technology (ART) procedures. However, the mechanisms leading to cell fragmentation remain largely unknown. Here, light sheet microscopy imaging of mouse embryos reveals that inefficient chromosome separation due to spindle defects, caused by dysfunctional molecular motors Myo1c or dynein, leads to fragmentation during mitosis. Extended exposure of the cell cortex to chromosomes locally triggers actomyosin contractility and pinches off cell fragments. This process is reminiscent of meiosis, during which small GTPase-mediated signals from chromosomes coordinate polar body extrusion (PBE) by actomyosin contraction. By interfering with the signals driving PBE, we find that this meiotic signaling pathway remains active during cleavage stages and is both required and sufficient to trigger fragmentation. Together, we find that fragmentation happens in mitosis after ectopic activation of actomyosin contractility by signals emanating from DNA, similar to those observed during meiosis. Our study uncovers the mechanisms underlying fragmentation in preimplantation embryos and, more generally, offers insight into the regulation of mitosis during the maternal-zygotic transition.


Assuntos
Actomiosina , Corpos Polares , Humanos , Animais , Camundongos , Corpos Polares/metabolismo , Actomiosina/metabolismo , Blastocisto , Cromossomos , Meiose , Oócitos/metabolismo , Fuso Acromático/genética , Miosina Tipo I/genética , Miosina Tipo I/metabolismo
2.
Genes Cells ; 29(5): 380-396, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38454557

RESUMO

Left-right (LR) asymmetry is crucial for animal development, particularly in Drosophila where LR-asymmetric morphogenesis of organs hinges on cellular-level chirality, termed cell chirality. In this species, two class I myosins, Myosin1D (Myo1D), and Myosin1C (Myo1C), respectively determine dextral (wild type) and sinistral (mirror image) cell chirality. Previous studies demonstrated Myo1D's ability to propel F-actin in leftward circles during in vitro gliding assays, suggesting its mechanochemical role in defining dextral chirality. Conversely, Myo1C propels F-actin without exhibiting LR-directional preference in this assay, suggesting at other properties governing sinistral chirality. Given the interaction of Myo1D and Myo1C with the membrane, we hypothesized that differences in their membrane behaviors might be critical in dictating their dextral or sinistral activities. In this study, employing single-molecule imaging analyses, we investigated the dynamic behaviors of Myo1D and Myo1C on the plasma membrane. Our findings revealed that Myo1C exhibits a significantly greater proportion of slow-diffusing population compared to Myo1D. Importantly, this characteristic was contingent upon both head and tail domains of Myo1C. The distinct diffusion patterns of Myo1D and Myo1C did not exert mutual influence on each other. This divergence in membrane diffusion between Myo1D and Myo1C may be crucial for dictating cell and organ chirality.


Assuntos
Membrana Celular , Proteínas de Drosophila , Macrófagos , Miosina Tipo I , Animais , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Miosina Tipo I/metabolismo , Miosina Tipo I/genética , Macrófagos/metabolismo , Drosophila melanogaster/metabolismo , Actinas/metabolismo , Imagem Individual de Molécula , Drosophila/metabolismo
3.
J Biol Chem ; 299(8): 104961, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37380077

RESUMO

Myosin-1D (myo1D) is important for Drosophila left-right asymmetry, and its effects are modulated by myosin-1C (myo1C). De novo expression of these myosins in nonchiral Drosophila tissues promotes cell and tissue chirality, with handedness depending on the paralog expressed. Remarkably, the identity of the motor domain determines the direction of organ chirality, rather than the regulatory or tail domains. Myo1D, but not myo1C, propels actin filaments in leftward circles in in vitro experiments, but it is not known if this property contributes to establishing cell and organ chirality. To further explore if there are differences in the mechanochemistry of these motors, we determined the ATPase mechanisms of myo1C and myo1D. We found that myo1D has a 12.5-fold higher actin-activated steady-state ATPase rate, and transient kinetic experiments revealed myo1D has an 8-fold higher MgADP release rate compared to myo1C. Actin-activated phosphate release is rate limiting for myo1C, whereas MgADP release is the rate-limiting step for myo1D. Notably, both myosins have among the tightest MgADP affinities measured for any myosin. Consistent with ATPase kinetics, myo1D propels actin filaments at higher speeds compared to myo1C in in vitro gliding assays. Finally, we tested the ability of both paralogs to transport 50 nm unilamellar vesicles along immobilized actin filaments and found robust transport by myo1D and actin binding but no transport by myo1C. Our findings support a model where myo1C is a slow transporter with long-lived actin attachments, whereas myo1D has kinetic properties associated with a transport motor.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Lateralidade Funcional , Miosina Tipo I , Animais , Actinas/metabolismo , Cinética , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Domínios Proteicos , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/enzimologia
4.
Eur J Immunol ; 53(3): e2250147, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36541400

RESUMO

VAV1-MYO1F is a recently identified gain-of-function fusion protein of the proto-oncogene Vav guanine nucleotide exchange factor 1 (VAV1) that is recurrently detected in T-cell non-Hodgkin's lymphoma (T-NHL) patients. However, the pathophysiological functions of VAV1-MYO1F in lymphomagenesis are insufficiently defined. Therefore, we generated transgenic mouse models to conditionally express VAV1-MYO1F in T-cells in vivo. We demonstrate that VAV1-MYO1F triggers cell autonomous activation of T-cell signaling with an activation of the ERK, JNK, and AKT pathways. VAV1-MYO1F expression induces a T-cell activation phenotype with high surface expression of CD25, ICOS, CD44, PD-1, and decreased CD62L as well as aberrant T-cell differentiation, proliferation, and neoplastic transformation. Consequently, the VAV1-MYO1F expressing T-cells induce a malignant T lymphoproliferative disease with 100% penetrance in vivo that mimics key aspects of human peripheral T-cell lymphoma. These results demonstrate that the human T-cell oncogene VAV1-MYO1F is sufficient to trigger oncogenic T-cell signaling and neoplastic transformation, and moreover, it provides a new clinically relevant mouse model to explore the pathogenesis of and treatment concepts for human T-cell lymphoma.


Assuntos
Linfoma de Células T Periférico , Proteínas Proto-Oncogênicas c-vav , Camundongos , Humanos , Animais , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Linfoma de Células T Periférico/genética , Transdução de Sinais , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Camundongos Transgênicos , Oncogenes , Miosina Tipo I/genética , Miosina Tipo I/metabolismo
5.
Brain Behav Immun ; 119: 750-766, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38710336

RESUMO

Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.


Assuntos
Dor Crônica , Disbiose , Gânglios Espinais , Microbioma Gastrointestinal , Camundongos Knockout , Miosina Tipo I , Animais , Feminino , Masculino , Camundongos , Dor Crônica/metabolismo , Dor Crônica/microbiologia , Disbiose/metabolismo , Gânglios Espinais/metabolismo , Microbioma Gastrointestinal/fisiologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Miosina Tipo I/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34301894

RESUMO

Opportunistic fungal infections have become one of the leading causes of death among immunocompromised patients, resulting in an estimated 1.5 million deaths each year worldwide. The molecular mechanisms that promote host defense against fungal infections remain elusive. Here, we find that Myosin IF (MYO1F), an unconventional myosin, promotes the expression of genes that are critical for antifungal innate immune signaling and proinflammatory responses. Mechanistically, MYO1F is required for dectin-induced α-tubulin acetylation, acting as an adaptor that recruits both the adaptor AP2A1 and α-tubulin N-acetyltransferase 1 to α-tubulin; in turn, these events control the membrane-to-cytoplasm trafficking of spleen tyrosine kinase and caspase recruitment domain-containing protein 9 Myo1f-deficient mice are more susceptible than their wild-type counterparts to the lethal sequelae of systemic infection with Candida albicans Notably, administration of Sirt2 deacetylase inhibitors, namely AGK2, AK-1, or AK-7, significantly increases the dectin-induced expression of proinflammatory genes in mouse bone marrow-derived macrophages and microglia, thereby protecting mice from both systemic and central nervous system C. albicans infections. AGK2 also promotes proinflammatory gene expression in human peripheral blood mononuclear cells after Dectin stimulation. Taken together, our findings describe a key role for MYO1F in promoting antifungal immunity by regulating the acetylation of α-tubulin and microtubules, and our findings suggest that Sirt2 deacetylase inhibitors may be developed as potential drugs for the treatment of fungal infections.


Assuntos
Candida albicans/fisiologia , Candidíase/imunologia , Imunidade Inata/imunologia , Leucócitos Mononucleares/imunologia , Microtúbulos/imunologia , Miosina Tipo I/metabolismo , Miosina Tipo I/fisiologia , Acetilação , Animais , Antifúngicos/farmacologia , Candidíase/tratamento farmacológico , Candidíase/metabolismo , Candidíase/microbiologia , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Microtúbulos/microbiologia , Miosina Tipo I/genética , Transdução de Sinais
7.
Development ; 147(3)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-31964776

RESUMO

Directional cell intercalations of epithelial cells during gastrulation has, in several organisms, been shown to be associated with a planar cell polarity in the organisation of the actin-myosin cytoskeleton and is postulated to reflect directional tension that drives oriented cell intercalations. We have characterised and applied a recently introduced non-destructive optical manipulation technique to measure the tension in individual epithelial cell junctions of cells in various locations and orientations in the epiblast of chick embryos in the early stages of primitive streak formation. Junctional tension of mesendoderm precursors in the epiblast is higher in junctions oriented in the direction of intercalation than in junctions oriented perpendicular to the direction of intercalation and higher than in junctions of other cells in the epiblast. The kinetic data fit best with a simple viscoelastic Maxwell model, and we find that junctional tension, and to a lesser extent viscoelastic relaxation time, are dependent on myosin activity.


Assuntos
Células Epiteliais/metabolismo , Gastrulação/fisiologia , Junções Intercelulares/metabolismo , Pinças Ópticas , Linha Primitiva/crescimento & desenvolvimento , Animais , Animais Geneticamente Modificados , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Embrião de Galinha , Gástrula/metabolismo , Camadas Germinativas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hidrocarbonetos Clorados/farmacologia , Microscopia de Fluorescência/métodos , Miosina Tipo I/antagonistas & inibidores , Miosina Tipo I/metabolismo , Miosina Tipo II/antagonistas & inibidores , Miosina Tipo II/metabolismo , Pirróis/farmacologia , Transdução de Sinais/fisiologia
8.
J Immunol ; 206(10): 2277-2289, 2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-33941653

RESUMO

The activation and degranulation of mast cells is critical in the pathogenesis of allergic inflammation and modulation of inflammation. Recently, we demonstrated that the unconventional long-tailed myosin, MYO1F, localizes with cortical F-actin and mediates adhesion and migration of mast cells. In this study, we show that knockdown of MYO1F by short hairpin RNA reduces human mast cell degranulation induced by both IgE crosslinking and by stimulation of the Mas-related G protein-coupled receptor X2 (MRGPRX2), which has been associated with allergic and pseudoallergic drug reactions, respectively. Defective degranulation was accompanied by a reduced reassembly of the cortical actin ring after activation but reversed by inhibition of actin polymerization. Our data show that MYO1F is required for full Cdc42 GTPase activation, a critical step in exocytosis. Furthermore, MYO1F knockdown resulted in less granule localization in the cell membrane and fewer fissioned mitochondria along with deficient mitochondria translocation to exocytic sites. Consistent with that, AKT and DRP1 phosphorylation are diminished in MYO1F knockdown cells. Altogether, our data point to MYO1F as an important regulator of mast cell degranulation by contributing to the dynamics of the cortical actin ring and the distribution of both the secretory granules and mitochondria.


Assuntos
Degranulação Celular/genética , Imunoglobulina E/metabolismo , Mastócitos/imunologia , Miosina Tipo I/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Doadores de Sangue , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Mitocôndrias/metabolismo , Miosina Tipo I/genética , Polimerização , Vesículas Secretórias/metabolismo , Transdução de Sinais/genética
9.
Pediatr Nephrol ; 38(2): 439-449, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35723736

RESUMO

BACKGROUND: Pathogenic mutations in the non-muscle single-headed myosin, myosin 1E (Myo1e), are a rare cause of pediatric focal segmental glomerulosclerosis (FSGS). These mutations are biallelic, to date only reported as homozygous variants in consanguineous families. Myo1e regulates the actin cytoskeleton dynamics and cell adhesion, which are especially important for podocyte functions. METHODS: DNA and RNA sequencing were used to identify novel MYO1E variants associated with FSGS. We studied the effects of these variants on the localization of Myo1e in kidney sections. We then analyzed the clinical and histological observations of all known pathogenic MYO1E variants. RESULTS: We identified a patient compound heterozygote for two novel variants in MYO1E and a patient homozygous for a deletion of exon 19. Computer modeling predicted these variants to be disruptive. In both patients, Myo1e was mislocalized. As a rule, pathogenic MYO1E variants map to the Myo1e motor and neck domain and are most often associated with steroid-resistant nephrotic syndrome in children 1-11 years of age, leading to kidney failure in 4-10 years in a subset of patients. The ultrastructural features are the podocyte damage and striking diffuse and global Alport-like glomerular basement membrane (GBM) abnormalities. CONCLUSIONS: We hypothesize that MYO1E mutations lead to disruption of the function of podocyte contractile actin cables resulting in abnormalities of the podocytes and the GBM and dysfunction of the glomerular filtration barrier. The characteristic clinicopathological data can help to tentatively differentiate this condition from other genetic podocytopathies and Alport syndrome until genetic testing is done. A higher resolution version of the Graphical abstract is available as Supplementary information.


Assuntos
Glomerulosclerose Segmentar e Focal , Nefrite Hereditária , Podócitos , Humanos , Membrana Basal Glomerular/patologia , Glomerulosclerose Segmentar e Focal/patologia , Mutação , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Nefrite Hereditária/genética , Fenótipo , Podócitos/patologia , Proteinúria/complicações
10.
Adv Exp Med Biol ; 1415: 499-505, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440078

RESUMO

Rods and cones are photoreceptor neurons in the retina that are required for visual sensation in vertebrates, where proper protein localization and compartmentalization are critical for phototransduction and visual function. In human retinal diseases, improper protein transport to the outer segment (OS) or mislocalization of proteins to the inner segment (IS) could lead to impaired visual responses and photoreceptor cell degeneration, causing a loss of visual function. We showed involvement of an unconventional motor protein, MYO1C, in the proper localization of rhodopsin to the OS, where loss of MYO1C in a mammalian model caused mislocalization of rhodopsin to IS and cell bodies, leading to progressively severe retinal phenotypes. In this study, using modeling and docking analysis, we aimed to identify the protein-protein interaction sites between MYO1C and Rhodopsin to establish a hypothesis that a physical interaction between these proteins is necessary for the proper trafficking of rhodopsin and visual function.


Assuntos
Retina , Rodopsina , Animais , Humanos , Rodopsina/genética , Rodopsina/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Transporte Proteico/fisiologia , Mamíferos/metabolismo , Miosina Tipo I/metabolismo
11.
J Am Soc Nephrol ; 33(11): 1989-2007, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36316095

RESUMO

BACKGROUND: Myo1e is a nonmuscle motor protein enriched in podocytes. Mutations in MYO1E are associated with steroid-resistant nephrotic syndrome (SRNS). Most of the MYO1E variants identified by genomic sequencing have not been functionally characterized. Here, we set out to analyze two mutations in the Myo1e motor domain, T119I and D388H, which were selected on the basis of protein sequence conservation. METHODS: EGFP-tagged human Myo1e constructs were delivered into the Myo1e-KO mouse podocyte-derived cells via adenoviral infection to analyze Myo1e protein stability, Myo1e localization, and clathrin-dependent endocytosis, which is known to involve Myo1e activity. Furthermore, truncated Myo1e constructs were expressed using the baculovirus expression system and used to measure Myo1e ATPase and motor activity in vitro. RESULTS: Both mutants were expressed as full-length proteins in the Myo1e-KO cells. However, unlike wild-type (WT) Myo1e, the T119I variant was not enriched at the cell junctions or clathrin-coated vesicles (CCVs). In contrast, D388H variant localization was similar to that of WT. The rate of dissociation of the D388H variant from cell-cell junctions and CCVs was decreased, suggesting this mutation affects Myo1e interactions with binding partners. ATPase activity and ability to translocate actin filaments were drastically reduced for the D388H mutant, supporting findings from cell-based experiments. CONCLUSIONS: T119I and D388H mutations are deleterious to Myo1e functions. The experimental approaches used in this study can be applied to future characterization of novel MYO1E variants associated with SRNS.


Assuntos
Miosina Tipo I , Síndrome Nefrótica , Animais , Humanos , Camundongos , Mutação , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Síndrome Nefrótica/genética , Esteroides
12.
J Biol Chem ; 296: 100128, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33257319

RESUMO

Myosin-1C is a single-headed, short-tailed member of the myosin class I subfamily that supports a variety of actin-based functions in the cytosol and nucleus. In vertebrates, alternative splicing of the MYO1C gene leads to the production of three isoforms, myosin-1C0, myosin-1C16, and myosin-1C35, that carry N-terminal extensions of different lengths. However, it is not clear how these extensions affect the chemomechanical coupling of human myosin-1C isoforms. Here, we report on the motor activity of the different myosin-1C isoforms measuring the unloaded velocities of constructs lacking the C-terminal lipid-binding domain on nitrocellulose-coated glass surfaces and full-length constructs on reconstituted, supported lipid bilayers. The higher yields of purified proteins obtained with constructs lacking the lipid-binding domain allowed a detailed characterization of the individual kinetic steps of human myosin-1C isoforms in their productive interaction with nucleotides and filamentous actin. Isoform-specific differences include 18-fold changes in the maximum power output per myosin-1C motor and 4-fold changes in the velocity and the resistive force at which maximum power output occurs. Our results support a model in which the isoform-specific N-terminal extensions affect chemomechanical coupling by combined steric and allosteric effects, thereby reducing both the length of the working stroke and the rate of ADP release in the absence of external loads by a factor of 2 for myosin-1C35. As the large change in maximum power output shows, the functional differences between the isoforms are further amplified by the presence of external loads.


Assuntos
Actinas/metabolismo , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Nucleotídeos/metabolismo , Fenômenos Biomecânicos , Humanos , Cinética , Ligação Proteica , Isoformas de Proteínas
13.
J Cell Physiol ; 237(9): 3671-3686, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35861939

RESUMO

Myosin-related proteins play an important role in cancer progression. However, the clinical significance, biological functions, and mechanisms of myosin 1B (MYO1B), in esophageal squamous cell carcinoma (ESCC) remain unclear. The clinical relevance of MYO1B, SNAI2, and cyclin D1 in ESCC was determined by immunohistochemistry, Oncomine, and GEPIA databases. The oncogenic roles of MYO1B were determined by CCK8, colony formation assays, wound healing, and Transwell assay. MYO1B, SNAI2, and cyclin D1 at mRNA and protein levels in ESCC cells were detected by qPCR and Western blot analysis. In our study, we found that MYO1B expression was increased in ESCC tissue samples and correlated with tumor stage, TNM stage, and poor outcomes. Functional assays indicated that depletion of MYO1B impaired oncogenesis, and enhanced chemosensitivity in ESCC. Bioinformatic analysis and mechanistic studies illustrated that SNAI2 was a key downstream effector of MYO1B. Suppression of MYO1B downregulated expression of SNAI2, thereby inhibiting the SNAI2/cyclin D1 pathway. Furthermore, a selective inhibitor of cyclin D1 activation reversed siMYO1B cells overexpressing SNAI2-elicited aggressive phenotypes of ESCC cells. MYO1B positively correlated with SNAI2 and cyclin D1 in ESCC samples, and higher SNAI2 expression was also associated with poor prognosis in ESCC patients. Our finding demonstrated that MYO1B activates the SNAI2/cyclin D1 pathway to drive tumorigenesis and cisplatin cytotoxicity in ESCC, indicating that MYO1B is a potential therapeutic target for patients with ESCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Carcinogênese/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/patologia , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Miosinas/metabolismo , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo
14.
J Cell Sci ; 133(5)2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-31964710

RESUMO

The inclusion of lymphocytes in high endothelial venules and their migration to the lymph nodes are critical steps in the immune response. Cell migration is regulated by the actin cytoskeleton and myosins. Myo1e is a long-tailed class I myosin and is highly expressed in B cells, which have not been studied in the context of cell migration. By using intravital microscopy in an in vivo model and performing in vitro experiments, we studied the relevance of Myo1e for the adhesion and inclusion of activated B cells in high endothelial venules. We observed reduced expression of integrins and F-actin in the membrane protrusions of B lymphocytes, which might be explained by deficiencies in vesicular trafficking. Interestingly, the lack of Myo1e reduced the phosphorylation of focal adhesion kinase (FAK; also known as PTK2), AKT (also known as AKT1) and RAC-1, disturbing the FAK-PI3K-RAC-1 signaling pathway. Taken together, our results indicate a critical role of Myo1e in the mechanism of B-cell adhesion and migration.


Assuntos
Miosina Tipo I , Miosinas , Actinas/metabolismo , Linfócitos B/metabolismo , Movimento Celular , Proteína-Tirosina Quinases de Adesão Focal , Linfonodos/metabolismo , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Miosinas/genética , Miosinas/metabolismo , Fosforilação
15.
PLoS Pathog ; 16(3): e1008323, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32163521

RESUMO

Fusarium is a genus of filamentous fungi that includes species that cause devastating diseases in major staple crops, such as wheat, maize, rice, and barley, resulting in severe yield losses and mycotoxin contamination of infected grains. Phenamacril is a novel fungicide that is considered environmentally benign due to its exceptional specificity; it inhibits the ATPase activity of the sole class I myosin of only a subset of Fusarium species including the major plant pathogens F. graminearum, F. asiaticum and F. fujikuroi. To understand the underlying mechanisms of inhibition, species specificity, and resistance mutations, we have determined the crystal structure of phenamacril-bound F. graminearum myosin I. Phenamacril binds in the actin-binding cleft in a new allosteric pocket that contains the central residue of the regulatory Switch 2 loop and that is collapsed in the structure of a myosin with closed actin-binding cleft, suggesting that pocket occupancy blocks cleft closure. We have further identified a single, transferable phenamacril-binding residue found exclusively in phenamacril-sensitive myosins to confer phenamacril selectivity.


Assuntos
Cianoacrilatos/química , Proteínas Fúngicas/química , Fungicidas Industriais/química , Fusarium/enzimologia , Miosina Tipo I/química , Cianoacrilatos/farmacologia , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Fungicidas Industriais/farmacologia , Fusarium/química , Fusarium/efeitos dos fármacos , Fusarium/genética , Miosina Tipo I/genética , Miosina Tipo I/metabolismo , Doenças das Plantas/microbiologia , Triticum/microbiologia , Zea mays/microbiologia
16.
FASEB J ; 35(2): e21268, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33470457

RESUMO

Several cytoskeleton-associated proteins and signaling pathways work in concert to regulate actin cytoskeleton remodeling, cell adhesion, and migration. Although the leukocyte-specific protein 1 (LSP1) has been shown to interact with the actin cytoskeleton, its function in the regulation of actin cytoskeleton dynamics is, as yet, not fully understood. We have recently demonstrated that the bimolecular complex between LSP1 and myosin1e controls actin cytoskeleton remodeling during phagocytosis. In this study, we show that LSP1 downregulation severely impairs cell migration, lamellipodia formation, and focal adhesion dynamics in macrophages. Inhibition of the interaction between LSP1 and myosin1e also impairs these processes resulting in poorly motile cells, which are characterized by few and small lamellipodia. Furthermore, cells in which LSP1-myosin1e interaction is inhibited are typically associated with inefficient focal adhesion turnover. Collectively, our findings show that the LSP1-myosin1e bimolecular complex plays a pivotal role in the regulation of actin cytoskeleton remodeling and focal adhesion dynamics required for cell migration.


Assuntos
Adesão Celular , Movimento Celular , Macrófagos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miosina Tipo I/metabolismo , Animais , Linhagem Celular , Matriz Extracelular/metabolismo , Macrófagos/fisiologia , Camundongos , Ligação Proteica , Pseudópodes/metabolismo
17.
J Biol Chem ; 295(12): 3757-3758, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32198186

RESUMO

Insulin-stimulated trafficking of GLUT4 requires the myosin motor Myo1C and signaling adaptor 14-3-3ß. Originally, it was thought that 14-3-3ß promotes GLUT4 transport by binding the Myo1C lever arm and activating the Myo1C motor. New work by Ji and Ostap using in vitro assays reveals that 14-3-3ß binding actually inhibits Myo1C motility, prompting reconsideration of the functional relationship between 14-3-3ß and Myo1C and the regulatory potential of atypical light chains.


Assuntos
Miosina Tipo I , Miosinas , Insulina , Miosina Tipo I/metabolismo , Miosinas/genética , Miosinas/metabolismo , Fosforilação , Transporte Proteico
18.
J Biol Chem ; 295(12): 3749-3756, 2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-31811090

RESUMO

Myosin-IC (Myo1c) has been proposed to function in delivery of glucose transporter type 4 (GLUT4)-containing vesicles to the plasma membrane in response to insulin stimulation. Current evidence suggests that, upon insulin stimulation, Myo1c is phosphorylated at Ser701, leading to binding of the signaling protein 14-3-3ß. Biochemical and functional details of the Myo1c-14-3-3ß interaction have yet to be described. Using recombinantly expressed proteins and mass spectrometry-based analyses to monitor Myo1c phosphorylation, along with pulldown, fluorescence binding, and additional biochemical assays, we show here that 14-3-3ß is a dimer and, consistent with previous work, that it binds to Myo1c in the presence of calcium. This interaction was associated with dissociation of calmodulin (CaM) from the IQ motif in Myo1c. Surprisingly, we found that 14-3-3ß binds to Myo1c independent of Ser701 phosphorylation in vitro Additionally, in contrast to previous reports, we did not observe Myo1c Ser701 phosphorylation by Ca2+/CaM-dependent protein kinase II (CaMKII), although CaMKII phosphorylated four other Myo1c sites. The presence of 14-3-3ß had little effect on the actin-activated ATPase or motile activities of Myo1c. Given these results, it is unlikely that 14-3-3ß acts as a cargo adaptor for Myo1c-powered transport; rather, we propose that 14-3-3ß binds Myo1c in the presence of calcium and stabilizes the calmodulin-dissociated, nonmotile myosin.


Assuntos
Proteínas 14-3-3/metabolismo , Miosina Tipo I/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/genética , Motivos de Aminoácidos , Cálcio/química , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calmodulina/metabolismo , Dimerização , Ácido Egtázico/química , Humanos , Espectrometria de Massas , Miosina Tipo I/química , Miosina Tipo I/genética , Fosforilação , Ligação Proteica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Ultracentrifugação
19.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1044-G1053, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33908271

RESUMO

Myosin 1c (Myo1c) is an unconventional myosin that modulates signaling pathways involved in tissue injury and repair. In this study, we observed that Myo1c expression is significantly upregulated in human chronic liver disease such as nonalcoholic steatohepatitis (NASH) and in animal models of liver fibrosis. High throughput data from the GEO-database identified similar Myo1c upregulation in mice and human liver fibrosis. Notably, transforming growth factor-ß1 (TGF-ß1) stimulation to hepatic stellate cells (HSCs), the liver pericyte and key cell type responsible for the deposition of extracellular matrix, upregulates Myo1c expression, whereas genetic depletion or pharmacological inhibition of Myo1c blunted TGF-ß-induced fibrogenic responses, resulting in repression of α-smooth muscle actin (α-SMA) and collagen type I α 1 chain (Col1α1) mRNA. Myo1c deletion also decreased fibrogenic processes such as cell proliferation, wound healing response, and contractility when compared with vehicle-treated HSCs. Importantly, phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) and mothers against decapentaplegic homolog 3 (SMAD3) were significantly blunted upon Myo1c inhibition in GRX cells as well as Myo1c knockout (Myo1c-KO) mouse embryonic fibroblasts (MEFs) upon TGF-ß stimulation. Using the genetic Myo1c-KO mice, we confirmed that Myo1c is critical for fibrogenesis, as Myo1c-KO mice were resistant to carbon tetrachloride (CCl4)-induced liver fibrosis. Histological and immunostaining analysis of liver sections showed that deposition of collagen fibers and α-SMA expression were significantly reduced in Myo1c-KO mice upon liver injury. Collectively, these results demonstrate that Myo1c mediates hepatic fibrogenesis by modulating TGF-ß signaling and suggest that inhibiting this process may have clinical application in treating liver fibrosis.NEW & NOTEWORTHY The incidences of liver fibrosis are growing at a rapid pace and have become one of the leading causes of end-stage liver disease. Although TGF-ß1 is known to play a prominent role in transforming cells to produce excessive extracellular matrix that lead to hepatic fibrosis, the therapies targeting TGF-ß1 have achieved very limited clinical impact. This study highlights motor protein myosin-1c-mediated mechanisms that serve as novel regulators of TGF-ß1 signaling and fibrosis.


Assuntos
Fibroblastos/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Miosina Tipo I/metabolismo , Animais , Cadeia alfa 1 do Colágeno Tipo I , Fibroblastos/patologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Camundongos , Miosina Tipo I/genética , Fosforilação , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
20.
J Cell Sci ; 132(8)2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30872458

RESUMO

In this study, we aimed to identify the myosin motor proteins that control trafficking at the Golgi complex. In addition to the known Golgi-associated myosins MYO6, MYO18A and MYH9 (myosin IIA), we identified MYO1C as a novel player at the Golgi in a human cell line. We demonstrate that depletion of MYO1C induces Golgi complex fragmentation and decompaction. MYO1C accumulates at dynamic structures around the Golgi complex that colocalize with Golgi-associated actin dots. MYO1C depletion leads to loss of cellular F-actin, and Golgi complex decompaction is also observed after inhibition or loss of the actin-related protein 2/3 complex, Arp2/3 (also known as ARPC). We show that the functional consequence of MYO1C depletion is a delay in the arrival of incoming transport carriers, both from the anterograde and retrograde routes. We propose that MYO1C stabilizes actin at the Golgi complex, facilitating the arrival of incoming transport carriers at the Golgi.This article has an associated First Person interview with the first author of the paper.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Complexo de Golgi/metabolismo , Miosina Tipo I/metabolismo , Linhagem Celular , Movimento Celular , Humanos , Miosina Tipo I/genética , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA