Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 338
Filtrar
1.
Nat Cell Biol ; 26(7): 1062-1076, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38951708

RESUMEN

Ras has been extensively studied as a promoter of cell proliferation, whereas few studies have explored its role in migration. To investigate the direct and immediate effects of Ras activity on cell motility or polarity, we focused on RasGAPs, C2GAPB in Dictyostelium amoebae and RASAL3 in HL-60 neutrophils and macrophages. In both cellular systems, optically recruiting the respective RasGAP to the cell front extinguished pre-existing protrusions and changed migration direction. However, when these respective RasGAPs were recruited uniformly to the membrane, cells polarized and moved more rapidly, whereas targeting to the back exaggerated these effects. These unexpected outcomes of attenuating Ras activity naturally had strong, context-dependent consequences for chemotaxis. The RasGAP-mediated polarization depended critically on myosin II activity and commenced with contraction at the cell rear, followed by sustained mTORC2-dependent actin polymerization at the front. These experimental results were captured by computational simulations in which Ras levels control front- and back-promoting feedback loops. The discovery that inhibiting Ras activity can produce counterintuitive effects on cell migration has important implications for future drug-design strategies targeting oncogenic Ras.


Asunto(s)
Actomiosina , Movimiento Celular , Polaridad Celular , Dictyostelium , Proteínas ras , Dictyostelium/metabolismo , Dictyostelium/genética , Células HL-60 , Actomiosina/metabolismo , Humanos , Proteínas ras/metabolismo , Proteínas ras/genética , Macrófagos/metabolismo , Miosina Tipo II/metabolismo , Miosina Tipo II/genética , Neutrófilos/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas Activadoras de ras GTPasa/genética , Animales , Quimiotaxis , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Actinas/metabolismo , Simulación por Computador , Ratones , Transducción de Señal
2.
Cell Mol Life Sci ; 81(1): 248, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38832964

RESUMEN

Contractile actomyosin bundles play crucial roles in various physiological processes, including cell migration, morphogenesis, and muscle contraction. The intricate assembly of actomyosin bundles involves the precise alignment and fusion of myosin II filaments, yet the underlying mechanisms and factors involved in these processes remain elusive. Our study reveals that LUZP1 plays a central role in orchestrating the maturation of thick actomyosin bundles. Loss of LUZP1 caused abnormal cell morphogenesis, migration, and the ability to exert forces on the environment. Importantly, knockout of LUZP1 results in significant defects in the concatenation and persistent association of myosin II filaments, severely impairing the assembly of myosin II stacks. The disruption of these processes in LUZP1 knockout cells provides mechanistic insights into the defective assembly of thick ventral stress fibers and the associated cellular contractility abnormalities. Overall, these results significantly contribute to our understanding of the molecular mechanism involved in actomyosin bundle formation and highlight the essential role of LUZP1 in this process.


Asunto(s)
Actomiosina , Movimiento Celular , Contracción Muscular , Miosina Tipo II , Humanos , Citoesqueleto de Actina/metabolismo , Actomiosina/metabolismo , Contracción Muscular/fisiología , Miosina Tipo II/metabolismo , Miosina Tipo II/genética
3.
PLoS Genet ; 20(6): e1011326, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38857279

RESUMEN

The development of ectodermal organs begins with the formation of a stratified epithelial placode that progressively invaginates into the underlying mesenchyme as the organ takes its shape. Signaling by secreted molecules is critical for epithelial morphogenesis, but how that information leads to cell rearrangement and tissue shape changes remains an open question. Using the mouse dentition as a model, we first establish that non-muscle myosin II is essential for dental epithelial invagination and show that it functions by promoting cell-cell adhesion and persistent convergent cell movements in the suprabasal layer. Shh signaling controls these processes by inducing myosin II activation via AKT. Pharmacological induction of AKT and myosin II can also rescue defects caused by the inhibition of Shh. Together, our results support a model in which the Shh signal is transmitted through myosin II to power effective cellular rearrangement for proper dental epithelial invagination.


Asunto(s)
Adhesión Celular , Movimiento Celular , Proteínas Hedgehog , Miosina Tipo II , Transducción de Señal , Animales , Ratones , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Adhesión Celular/genética , Miosina Tipo II/metabolismo , Miosina Tipo II/genética , Movimiento Celular/genética , Epitelio/metabolismo , Morfogénesis/genética , Diente/metabolismo , Diente/crecimiento & desarrollo , Células Epiteliales/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Regulación del Desarrollo de la Expresión Génica
4.
Cell Mol Life Sci ; 81(1): 195, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38653877

RESUMEN

The Notch pathway is an evolutionarily conserved signaling system that is intricately regulated at multiple levels and it influences different aspects of development. In an effort to identify novel components involved in Notch signaling and its regulation, we carried out protein interaction screens which identified non-muscle myosin II Zipper (Zip) as an interacting partner of Notch. Physical interaction between Notch and Zip was further validated by co-immunoprecipitation studies. Immunocytochemical analyses revealed that Notch and Zip co-localize within same cytoplasmic compartment. Different alleles of zip also showed strong genetic interactions with Notch pathway components. Downregulation of Zip resulted in wing phenotypes that were reminiscent of Notch loss-of-function phenotypes and a perturbed expression of Notch downstream targets, Cut and Deadpan. Further, synergistic interaction between Notch and Zip resulted in highly ectopic expression of these Notch targets. Activated Notch-induced tumorous phenotype of larval tissues was enhanced by over-expression of Zip. Notch-Zip synergy resulted in the activation of JNK pathway that consequently lead to MMP activation and proliferation. Taken together, our results suggest that Zip may play an important role in regulation of Notch signaling.


Asunto(s)
Proteínas de Drosophila , Proteínas de la Membrana , Cadenas Pesadas de Miosina , Receptores Notch , Transducción de Señal , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores Notch/metabolismo , Receptores Notch/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Alas de Animales/metabolismo , Alas de Animales/crecimiento & desarrollo , Drosophila/metabolismo , Drosophila/genética , Fenotipo , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Proliferación Celular , Miosina Tipo II/metabolismo , Miosina Tipo II/genética
5.
J Cell Biol ; 222(11)2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37843812

RESUMEN

During the second meiotic cell division, egg cells discard one set of chromatids to the polar body to produce a large haploid gamete. Meiotic spindle rotation is a critical step to ensure proper polar body extrusion. In this issue, Bourdais et al. (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202211029) have identified MRCKß as an essential kinase for efficient spindle rotation. MRCK activates cortical myosin II rings overlying the spindle to prevent the notoriously sticky interaction between the cell cortex and chromatin to facilitate spindle rotation. Furthermore, Bourdais et al. found that the same MRCK-myosin II pathway also operates in zygotes to promote parental genome unification.


Asunto(s)
Cromatina , Cromosomas , Cromatina/metabolismo , Rotación , Huso Acromático/metabolismo , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Oocitos/metabolismo , Meiosis
6.
Elife ; 122023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37724949

RESUMEN

Cell spreading and migration play central roles in many physiological and pathophysiological processes. We have previously shown that MFN2 regulates the migration of human neutrophil-like cells via suppressing Rac activation. Here, we show that in mouse embryonic fibroblasts, MFN2 suppresses RhoA activation and supports cell polarization. After initial spreading, the wild-type cells polarize and migrate, whereas the Mfn2-/- cells maintain a circular shape. Increased cytosolic Ca2+ resulting from the loss of Mfn2 is directly responsible for this phenotype, which can be rescued by expressing an artificial tether to bring mitochondria and endoplasmic reticulum to close vicinity. Elevated cytosolic Ca2+ activates Ca2+/calmodulin-dependent protein kinase II, RhoA, and myosin light-chain kinase, causing an overactivation of nonmuscle myosin II, leading to a formation of a prominent F-actin ring at the cell periphery and increased cell contractility. The peripheral actin band alters cell physics and is dependent on substrate rigidity. Our results provide a novel molecular basis to understand how MFN2 regulates distinct signaling pathways in different cells and tissue environments, which is instrumental in understanding and treating MFN2-related diseases.


Asunto(s)
Actinas , Fibroblastos , Animales , Humanos , Ratones , Actinas/metabolismo , Fibroblastos/metabolismo , Transducción de Señal , Retículo Endoplásmico/metabolismo , Miosina Tipo II/genética , Miosina Tipo II/metabolismo
7.
Elife ; 122023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36715100

RESUMEN

The actomyosin cytoskeleton is a crucial driver of morphogenesis. Yet how the behavior of large-scale cytoskeletal patterns in deforming tissues emerges from the interplay of geometry, genetics, and mechanics remains incompletely understood. Convergent extension in Drosophila melanogaster embryos provides the opportunity to establish a quantitative understanding of the dynamics of anisotropic non-muscle myosin II. Cell-scale analysis of protein localization in fixed embryos suggests that gene expression patterns govern myosin anisotropy via complex rules. However, technical limitations have impeded quantitative and dynamic studies of this process at the whole embryo level, leaving the role of geometry open. Here, we combine in toto live imaging with quantitative analysis of molecular dynamics to characterize the distribution of myosin anisotropy and the corresponding genetic patterning. We found pair rule gene expression continuously deformed, flowing with the tissue frame. In contrast, myosin anisotropy orientation remained approximately static and was only weakly deflected from the stationary dorsal-ventral axis of the embryo. We propose that myosin is recruited by a geometrically defined static source, potentially related to the embryo-scale epithelial tension, and account for transient deflections by cytoskeletal turnover and junction reorientation by flow. With only one parameter, this model quantitatively accounts for the time course of myosin anisotropy orientation in wild-type, twist, and even-skipped embryos, as well as embryos with perturbed egg geometry. Geometric patterning of the cytoskeleton suggests a simple physical strategy to ensure a robust flow and formation of shape.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Morfogénesis , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Miosinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Embrión no Mamífero/metabolismo
8.
J Cell Biol ; 222(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36416725

RESUMEN

Fibronectin (FN) is an essential structural and regulatory component of the extracellular matrix (ECM), and its binding to integrin receptors supports cell adhesion, migration, and signaling. Here, using live-cell microscopy of fibroblasts expressing FN tagged with a pH-sensitive fluorophore, we show that FN is secreted predominantly at the ventral surface of cells in an integrin-independent manner. Locally secreted FN then undergoes ß1 integrin-dependent fibrillogenesis. We find that the site of FN secretion is regulated by cell polarization, which occurs in bursts under stabilized lamellipodia at the leading edge. Moreover, analysis of FN secretion and focal adhesion dynamics suggest that focal adhesion formation precedes FN deposition and that deposition continues during focal adhesion disassembly. Lastly, we show that the polarized FN deposition in spreading and migrating cells requires both intact microtubules and myosin II-mediated contractility. Thus, while FN secretion does not require integrin binding, the site of exocytosis is regulated by membrane and cytoskeletal dynamics with secretion occurring after new adhesion formation.


Asunto(s)
Fibronectinas , Microtúbulos , Miosina Tipo II , Seudópodos , Proteínas del Citoesqueleto/metabolismo , Fibroblastos/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Integrinas/metabolismo , Microtúbulos/genética , Microtúbulos/metabolismo , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Seudópodos/genética , Seudópodos/metabolismo , Matriz Extracelular/metabolismo , Exocitosis
9.
J Cell Sci ; 136(5)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36052670

RESUMEN

In common with other actomyosin contractile cellular machineries, actin turnover is required for normal function of the cytokinetic contractile ring. Cofilin is an actin-binding protein contributing to turnover by severing actin filaments, required for cytokinesis by many organisms. In fission yeast cofilin mutants, contractile rings suffer bridging instabilities in which segments of the ring peel away from the plasma membrane, forming straight bridges whose ends remain attached to the membrane. The origin of bridging instability is unclear. Here, we used molecularly explicit simulations of contractile rings to examine the role of cofilin. Simulations reproduced the experimentally observed cycles of bridging and reassembly during constriction, and the occurrence of bridging in ring segments with low density of the myosin II protein Myo2. The lack of cofilin severing produced ∼2-fold longer filaments and, consequently, ∼2-fold higher ring tensions. Simulations identified bridging as originating in the boosted ring tension, which increased centripetal forces that detached actin from Myo2, which was anchoring actin to the membrane. Thus, cofilin serves a critical role in cytokinesis by providing protection from bridging, the principal structural threat to contractile rings.


Asunto(s)
Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Citoesqueleto de Actina/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Actomiosina/metabolismo , Citocinesis , Proteínas de Microfilamentos/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
10.
Curr Biol ; 32(12): 2704-2718.e6, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35594862

RESUMEN

Cancer cell migration during metastasis is mediated by a highly polarized cytoskeleton. MARK2 and its invertebrate homolog Par1B are kinases that regulate the microtubule cytoskeleton to mediate polarization of neurons in mammals and embryos in invertebrates. However, the role of MARK2 in cancer cell migration is unclear. Using osteosarcoma cells, we found that in addition to its known localizations on microtubules and the plasma membrane, MARK2 also associates with the actomyosin cytoskeleton and focal adhesions. Cells depleted of MARK proteins demonstrated that MARK2 promotes phosphorylation of both myosin II and the myosin phosphatase targeting subunit MYPT1 to synergistically drive myosin II contractility and stress fiber formation in cells. Studies with isolated proteins showed that MARK2 directly phosphorylates myosin II regulatory light chain, while its effects on MYPT1 phosphorylation are indirect. Using a mutant lacking the membrane-binding domain, we found that membrane association is required for focal adhesion targeting of MARK2, where it specifically enhances cell protrusion by promoting FAK phosphorylation and formation of focal adhesions oriented in the direction of migration to mediate directionally persistent cell motility. Together, our results define MARK2 as a master regulator of the actomyosin and microtubule cytoskeletal systems and focal adhesions to mediate directional cancer cell migration.


Asunto(s)
Actomiosina , Adhesiones Focales , Actomiosina/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Mamíferos , Cadenas Ligeras de Miosina/metabolismo , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Fosforilación
11.
Development ; 149(10)2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35451459

RESUMEN

Apical constriction, or a reduction in size of the apical domain, underlies many morphogenetic events during development. Actomyosin complexes play an essential role in apical constriction; however, the detailed analysis of molecular mechanisms is still pending. Here, we show that Lim domain only protein 7 (Lmo7), a multidomain adaptor at apical junctions, promotes apical constriction in the Xenopus superficial ectoderm, whereas apical domain size increases in Lmo7-depleted cells. Lmo7 is primarily localized at apical junctions and promotes the formation of the dense circumferential actomyosin belt. Strikingly, Lmo7 binds non-muscle myosin II (NMII) and recruits it to apical junctions and the apical cortex. This NMII recruitment is essential for Lmo7-mediated apical constriction. Lmo7 knockdown decreases NMIIA localization at apical junctions and delays neural tube closure in Xenopus embryos. Our findings suggest that Lmo7 serves as a scaffold that regulates actomyosin contractility and apical domain size.


Asunto(s)
Actomiosina , Ectodermo , Actomiosina/metabolismo , Animales , Ectodermo/metabolismo , Morfogénesis/fisiología , Cadenas Pesadas de Miosina , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Xenopus laevis/metabolismo
12.
Mol Biol Cell ; 33(6): ar51, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34613787

RESUMEN

Cytokinesis by animals, fungi, and amoebas depends on actomyosin contractile rings, which are stabilized by continuous turnover of actin filaments. Remarkably little is known about the amount of polymerized actin in contractile rings, so we used low concentrations of GFP-Lifeact to count total polymerized actin molecules in the contractile rings of live fission yeast cells. Contractile rings of wild-type cells accumulated polymerized actin molecules at 4900/min to a peak number of ∼198,000 followed by a loss of actin at 5400/min throughout ring constriction. In adf1-M3 mutant cells with cofilin that severs actin filaments poorly, contractile rings accumulated polymerized actin at twice the normal rate and eventually had almost twofold more actin along with a proportional increase in type II myosins Myo2, Myp2, and formin Cdc12. Although 30% of adf1-M3 mutant cells failed to constrict their rings fully, the rest lost actin from the rings at the wild-type rates. Mutations of type II myosins Myo2 and Myp2 reduced contractile ring actin filaments by half and slowed the rate of actin loss from the rings.


Asunto(s)
Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Citoesqueleto de Actina , Factores Despolimerizantes de la Actina , Actinas , Animales , Citocinesis/genética , Miopía , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Miosinas , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética
13.
Sci Rep ; 11(1): 22755, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815439

RESUMEN

Mitochondrial DNA (mtDNA) maintenance is essential to sustain a functionally healthy population of mitochondria within cells. Proper mtDNA replication and distribution within mitochondrial networks are essential to maintain mitochondrial homeostasis. However, the fundamental basis of mtDNA segregation and distribution within mitochondrial networks is still unclear. To address these questions, we developed an algorithm, Mitomate tracker to unravel the global distribution of nucleoids within mitochondria. Using this tool, we decipher the semi-regular spacing of nucleoids across mitochondrial networks. Furthermore, we show that mitochondrial fission actively regulates mtDNA distribution by controlling the distribution of nucleoids within mitochondrial networks. Specifically, we found that primary cells bearing disease-associated mutations in the fission proteins DRP1 and MYH14 show altered nucleoid distribution, and acute enrichment of enlarged nucleoids near the nucleus. Further analysis suggests that the altered nucleoid distribution observed in the fission mutants is the result of both changes in network structure and nucleoid density. Thus, our study provides novel insights into the role of mitochondria fission in nucleoid distribution and the understanding of diseases caused by fission defects.


Asunto(s)
Núcleo Celular/metabolismo , ADN Mitocondrial/metabolismo , Dinaminas/metabolismo , Homeostasis , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo II/metabolismo , Núcleo Celular/genética , Replicación del ADN , ADN Mitocondrial/genética , Dinaminas/genética , Humanos , Mitocondrias/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética
14.
Genes (Basel) ; 12(10)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34681017

RESUMEN

Variants in MYH14 are reported to cause autosomal dominant nonsyndromic hereditary hearing loss (ADNSHL), with 34 variants reported to cause hearing loss in various ethnic groups. However, the available information on prevalence, as well as with regard to clinical features, remains fragmentary. In this study, genetic screening for MYH14 variants was carried out using a large series of Japanese hearing-loss patients to reveal more detailed information. Massively parallel DNA sequencing of 68 target candidate genes was applied in 8074 unrelated Japanese hearing-loss patients (including 1336 with ADNSHL) to identify genomic variations responsible for hearing loss. We identified 11 families with 10 variants. The prevalence was found to be 0.14% (11/8074) among all hearing-loss patients and 0.82% (11/1336) among ADNSHL patients. Nine of the eleven variants identified were novel. The patients typically showed late-onset hearing loss arising later than 20 years of age (64.3%, 9/14) along with progressive (92.3%, 12/13), moderate (62.5%, 10/16), and flat-type hearing loss (68.8%, 11/16). We also confirmed progressive hearing loss in serial audiograms. The clinical information revealed by the present study will contribute to further diagnosis and management of MYH14-associated hearing loss.


Asunto(s)
Sordera/genética , Predisposición Genética a la Enfermedad , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Adolescente , Adulto , Secuencia de Aminoácidos/genética , Pueblo Asiatico , Sordera/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje , Análisis de Secuencia de ADN
15.
Elife ; 102021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34374341

RESUMEN

Nonmuscle myosin II (NM II) is an integral part of essential cellular processes, including adhesion and migration. Mammalian cells express up to three isoforms termed NM IIA, B, and C. We used U2OS cells to create CRISPR/Cas9-based knockouts of all three isoforms and analyzed the phenotypes on homogenously coated surfaces, in collagen gels, and on micropatterned substrates. In contrast to homogenously coated surfaces, a structured environment supports a cellular phenotype with invaginated actin arcs even in the absence of NM IIA-induced contractility. A quantitative shape analysis of cells on micropatterns combined with a scale-bridging mathematical model reveals that NM IIA is essential to build up cellular tension during initial stages of force generation, while NM IIB is necessary to elastically stabilize NM IIA-generated tension. A dynamic cell stretch/release experiment in a three-dimensional scaffold confirms these conclusions and in addition reveals a novel role for NM IIC, namely the ability to establish tensional homeostasis.


Asunto(s)
Elasticidad , Miosina Tipo II/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Movimiento Celular/fisiología , Homeostasis , Humanos , Modelos Teóricos , Miosina Tipo II/clasificación , Miosina Tipo II/genética , Miosina Tipo IIA no Muscular/genética , Miosina Tipo IIB no Muscular/genética , Isoformas de Proteínas
16.
J Am Heart Assoc ; 10(14): e020965, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34227390

RESUMEN

Background Although the roles of alpha-myosin heavy chain (α-MyHC) and beta-myosin heavy chain (ß-MyHC) proteins in cardiac contractility have long been appreciated, the biological contribution of another closely related sarcomeric myosin family member, MYH7b (myosin heavy chain 7b), has become a matter of debate. In mammals, MYH7b mRNA is transcribed but undergoes non-productive alternative splicing that prevents protein expression in a tissue-specific manner, including in the heart. However, several studies have recently linked MYH7b variants to different cardiomyopathies or have reported MYH7b protein expression in mammalian hearts. Methods and Results By analyzing mammalian cardiac transcriptome and proteome data, we show that the vast majority of MYH7b RNA is subject to exon skipping and cannot be translated into a functional myosin molecule. Notably, we discovered a lag in the removal of introns flanking the alternatively spliced exon, which could retain the non-coding RNA in the nucleus. This process could play a significant role in controlling MYH7b expression as well as the activity of other cardiac genes. Consistent with the negligible level of full-length protein coding mRNA, no MYH7b protein expression was detected in adult mouse, rat, and human hearts by Western blot analysis. Furthermore, proteome surveys including quantitative mass spectrometry analyses revealed only traces of cardiac MYH7b protein and even then, only in a subset of individual samples. Conclusions The comprehensive analysis presented here suggests that previous studies showing cardiac MYH7b protein expression were likely attributable to antibody cross-reactivity. More importantly, our data predict that the MYH7b disease-associated variants may operate through the alternately spliced RNA itself.


Asunto(s)
Cardiomiopatías/genética , Regulación de la Expresión Génica , Ventrículos Cardíacos/patología , Contracción Miocárdica/fisiología , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/genética , Miosina Tipo II/genética , Animales , Western Blotting , Cadáver , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Ventrículos Cardíacos/metabolismo , Humanos , Mamíferos , Ratones , Miocardio/patología , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/biosíntesis , Miosina Tipo II/biosíntesis , ARN/genética , ARN Mensajero/genética , Ratas
17.
Biophys J ; 120(19): 4214-4229, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34293302

RESUMEN

Distinct patterns of actomyosin contractility are often associated with particular epithelial tissue shape changes during development. For example, a planar-polarized pattern of myosin II localization regulated by Rho1 signaling during Drosophila body axis elongation is thought to drive cell behaviors that contribute to convergent extension. However, it is not well understood how specific aspects of a myosin pattern influence the multiple cell behaviors, including cell intercalation, cell shape changes, and apical cell area fluctuations, that simultaneously occur during morphogenesis. Here, we developed two optogenetic tools, optoGEF and optoGAP, to activate or deactivate Rho1 signaling, respectively. We used these tools to manipulate myosin patterns at the apical side of the germband epithelium during Drosophila axis elongation and analyzed the effects on contractile cell behaviors. We show that uniform activation or inactivation of Rho1 signaling across the apical surface of the germband is sufficient to disrupt the planar-polarized pattern of myosin at cell junctions on the timescale of 3-5 min, leading to distinct changes in junctional and medial myosin patterns in optoGEF and optoGAP embryos. These two perturbations to Rho1 activity both disrupt axis elongation and cell intercalation but have distinct effects on cell area fluctuations and cell packings that are linked with changes in the medial and junctional myosin pools. These studies demonstrate that acute optogenetic perturbations to Rho1 activity are sufficient to rapidly override the endogenous planar-polarized myosin pattern in the germband during axis elongation. Moreover, our results reveal that the levels of Rho1 activity and the balance between medial and junctional myosin play key roles not only in organizing the cell rearrangements that are known to directly contribute to axis elongation but also in regulating cell area fluctuations and cell packings, which have been proposed to be important factors influencing the mechanics of tissue deformation and flow.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Animales , Polaridad Celular , Proteínas de Drosophila/genética , Morfogénesis , Miosina Tipo II/genética , Optogenética
18.
Annu Rev Cell Dev Biol ; 37: 285-310, 2021 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-34314591

RESUMEN

Nonmuscle myosin II (NMII) is a multimeric protein complex that generates most mechanical force in eukaryotic cells. NMII function is controlled at three main levels. The first level includes events that trigger conformational changes that extend the complex to enable its assembly into filaments. The second level controls the ATPase activity of the complex and its binding to microfilaments in extended NMII filaments. The third level includes events that modulate the stability and contractility of the filaments. They all work in concert to finely control force generation inside cells. NMII is a common endpoint of mechanochemical signaling pathways that control cellular responses to physical and chemical extracellular cues. Specific phosphorylations modulate NMII activation in a context-dependent manner. A few kinases control these phosphorylations in a spatially, temporally, and lineage-restricted fashion, enabling functional adaptability to the cellular microenvironment. Here, we review mechanisms that control NMII activity in the context of cell migration and division.


Asunto(s)
Citoesqueleto , Miosina Tipo II , Citoesqueleto de Actina/metabolismo , Movimiento Celular/genética , Citoesqueleto/metabolismo , Miosina Tipo II/química , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Transducción de Señal
19.
Cells ; 10(5)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067877

RESUMEN

Wound repair of cell membranes is essential for cell survival. Myosin II contributes to wound pore closure by interacting with actin filaments in larger cells; however, its role in smaller cells is unclear. In this study, we observed wound repair in dividing cells for the first time. The cell membrane in the cleavage furrow, where myosin II localized, was wounded by laserporation. Upon wounding, actin transiently accumulated, and myosin II transiently disappeared from the wound site. Ca2+ influx from the external medium triggered both actin and myosin II dynamics. Inhibition of calmodulin reduced both actin and myosin II dynamics. The wound closure time in myosin II-null cells was the same as that in wild-type cells, suggesting that myosin II is not essential for wound repair. We also found that disassembly of myosin II filaments by phosphorylation did not contribute to their disappearance, indicating a novel mechanism for myosin II delocalization from the cortex. Furthermore, we observed that several furrow-localizing proteins such as GAPA, PakA, myosin heavy chain kinase C, PTEN, and dynamin disappeared upon wounding. Herein, we discuss the possible mechanisms of myosin dynamics during wound repair.


Asunto(s)
División Celular , Dictyostelium/metabolismo , Miosina Tipo II/metabolismo , Proteínas Protozoarias/metabolismo , Cicatrización de Heridas , Calcio/metabolismo , Señalización del Calcio , Dictyostelium/genética , Dictyostelium/crecimiento & desarrollo , Cinética , Microscopía Fluorescente , Microscopía por Video , Mutación , Miosina Tipo II/genética , Proteínas Protozoarias/genética , Imagen de Lapso de Tiempo
20.
Nat Cell Biol ; 23(7): 733-744, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34155381

RESUMEN

Intestinal organoids derived from single cells undergo complex crypt-villus patterning and morphogenesis. However, the nature and coordination of the underlying forces remains poorly characterized. Here, using light-sheet microscopy and large-scale imaging quantification, we demonstrate that crypt formation coincides with a stark reduction in lumen volume. We develop a 3D biophysical model to computationally screen different mechanical scenarios of crypt morphogenesis. Combining this with live-imaging data and multiple mechanical perturbations, we show that actomyosin-driven crypt apical contraction and villus basal tension work synergistically with lumen volume reduction to drive crypt morphogenesis, and demonstrate the existence of a critical point in differential tensions above which crypt morphology becomes robust to volume changes. Finally, we identified a sodium/glucose cotransporter that is specific to differentiated enterocytes that modulates lumen volume reduction through cell swelling in the villus region. Together, our study uncovers the cellular basis of how cell fate modulates osmotic and actomyosin forces to coordinate robust morphogenesis.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Mucosa Intestinal/fisiología , Mecanotransducción Celular , Osmorregulación , Células de Paneth/fisiología , Células Madre/fisiología , Animales , Movimiento Celular , Células Cultivadas , Simulación por Computador , Femenino , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía por Video , Modelos Biológicos , Morfogénesis , Miosina Tipo II/genética , Miosina Tipo II/metabolismo , Organoides , Presión Osmótica , Células de Paneth/metabolismo , Proteínas de Transporte de Sodio-Glucosa/genética , Proteínas de Transporte de Sodio-Glucosa/metabolismo , Células Madre/metabolismo , Estrés Mecánico , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...