Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37921850

RESUMO

Sarcomeres are the basic contractile units within cardiac myocytes, and the collective shortening of sarcomeres aligned along myofibrils generates the force driving the heartbeat. The alignment of the individual sarcomeres is important for proper force generation, and misaligned sarcomeres are associated with diseases, including cardiomyopathies and COVID-19. The actin bundling protein, α-actinin-2, localizes to the 'Z-Bodies" of sarcomere precursors and the 'Z-Lines' of sarcomeres, and has been used previously to assess sarcomere assembly and maintenance. Previous measurements of α-actinin-2 organization have been largely accomplished manually, which is time-consuming and has hampered research progress. Here, we introduce sarcApp, an image analysis tool that quantifies several components of the cardiac sarcomere and their alignment in muscle cells and tissue. We first developed sarcApp to utilize deep learning-based segmentation and real space quantification to measure α-actinin-2 structures and determine the organization of both precursors and sarcomeres/myofibrils. We then expanded sarcApp to analyze 'M-Lines' using the localization of myomesin and a protein that connects the Z-Lines to the M-Line (titin). sarcApp produces 33 distinct measurements per cell and 24 per myofibril that allow for precise quantification of changes in sarcomeres, myofibrils, and their precursors. We validated this system with perturbations to sarcomere assembly. We found perturbations that affected Z-Lines and M-Lines differently, suggesting that they may be regulated independently during sarcomere assembly.


Assuntos
Miócitos Cardíacos , Sarcômeros , Sarcômeros/metabolismo , Miócitos Cardíacos/metabolismo , Actinina/metabolismo , Miofibrilas/metabolismo , Conectina/metabolismo , Software
2.
bioRxiv ; 2023 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-36711995

RESUMO

Sarcomeres are the basic contractile units within cardiac myocytes, and the collective shortening of sarcomeres aligned along myofibrils generates the force driving the heartbeat. The alignment of the individual sarcomeres is important for proper force generation, and misaligned sarcomeres are associated with diseases including cardiomyopathies and COVID-19. The actin bundling protein, α-actinin-2, localizes to the "Z-Bodies" of sarcomere precursors and the "Z-Lines" of sarcomeres, and has been used previously to assess sarcomere assembly and maintenance. Previous measurements of α-actinin-2 organization have been largely accomplished manually, which is time-consuming and has hampered research progress. Here, we introduce sarcApp, an image analysis tool that quantifies several components of the cardiac sarcomere and their alignment in muscle cells and tissue. We first developed sarcApp to utilize deep learning-based segmentation and real space quantification to measure α-actinin-2 structures and determine the organization of both precursors and sarcomeres/myofibrils. We then expanded sarcApp to analyze "M-Lines" using the localization of myomesin and a protein that connects the Z-Lines to the M-Line (titin). sarcApp produces 33 distinct measurements per cell and 24 per myofibril that allow for precise quantification of changes in sarcomeres, myofibrils, and their precursors. We validated this system with perturbations to sarcomere assembly. We found perturbations that affected Z-Lines and M-Lines differently, suggesting that they may be regulated independently during sarcomere assembly.

3.
Nat Phys ; 18(9): 1112-1121, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37220497

RESUMO

Cell behaviour is affected by the physical forces and mechanical properties of the cells and of their microenvironment. The viscosity of extracellular fluid - a component of the cellular microenvironment - can vary by orders of magnitude, but its effect on cell behaviour remains largely unexplored. Using bio-compatible polymers to increase the viscosity of the culture medium, we characterize how viscosity affects cell behaviour. We find that multiple types of adherent cells respond in an unexpected but similar manner to elevated viscosity. In a highly viscous medium, cells double their spread area, exhibit increased focal adhesion formation and turnover, generate significantly greater traction forces, and migrate nearly two times faster. We observe that when cells are immersed in regular medium, these viscosity-dependent responses require an actively ruffling lamellipodium - a dynamic membrane structure at the front of the cell. We present evidence that cells utilize membrane ruffling to sense changes in extracellular fluid viscosity and to trigger adaptive responses.

4.
Nat Cell Biol ; 23(12): 1240-1254, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34887515

RESUMO

Extracellular vesicles and exomere nanoparticles are under intense investigation as sources of clinically relevant cargo. Here we report the discovery of a distinct extracellular nanoparticle, termed supermere. Supermeres are morphologically distinct from exomeres and display a markedly greater uptake in vivo compared with small extracellular vesicles and exomeres. The protein and RNA composition of supermeres differs from small extracellular vesicles and exomeres. Supermeres are highly enriched with cargo involved in multiple cancers (glycolytic enzymes, TGFBI, miR-1246, MET, GPC1 and AGO2), Alzheimer's disease (APP) and cardiovascular disease (ACE2, ACE and PCSK9). The majority of extracellular RNA is associated with supermeres rather than small extracellular vesicles and exomeres. Cancer-derived supermeres increase lactate secretion, transfer cetuximab resistance and decrease hepatic lipids and glycogen in vivo. This study identifies a distinct functional nanoparticle replete with potential circulating biomarkers and therapeutic targets for a host of human diseases.


Assuntos
Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo , Nanopartículas/metabolismo , Doença de Alzheimer/patologia , Enzima de Conversão de Angiotensina 2/metabolismo , Transporte Biológico/fisiologia , Biomarcadores/metabolismo , COVID-19/patologia , Doenças Cardiovasculares/patologia , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Células HeLa , Humanos , Ácido Láctico/metabolismo , MicroRNAs/genética , Nanopartículas/classificação , Neoplasias/patologia , Microambiente Tumoral
5.
Mol Biol Cell ; 32(20): br3, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34319762

RESUMO

Force generation by the molecular motor myosin II (MII) at the actin cortex is a universal feature of animal cells. Despite its central role in driving cell shape changes, the mechanisms underlying MII regulation at the actin cortex remain incompletely understood. Here we show that myosin light chain kinase (MLCK) promotes MII turnover at the mitotic cortex. Inhibition of MLCK resulted in an alteration of the relative levels of phosphorylated regulatory light chain (RLC), with MLCK preferentially creating a short-lived pRLC species and Rho-associated kinase (ROCK) preferentially creating a stable ppRLC species during metaphase. Slower turnover of MII and altered RLC homeostasis on MLCK inhibition correlated with increased cortex tension, driving increased membrane bleb initiation and growth, but reduced bleb retraction during mitosis. Taken together, we show that ROCK and MLCK play distinct roles at the actin cortex during mitosis; ROCK activity is required for recruitment of MII to the cortex, while MLCK activity promotes MII turnover. Our findings support the growing evidence that MII turnover is an essential dynamic process influencing the mechanical output of the actin cortex.


Assuntos
Actinas , Proteínas de Ligação ao Cálcio , Miosina Tipo II , Quinase de Cadeia Leve de Miosina , Humanos , Actinas/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/fisiologia , Divisão do Núcleo Celular , Proteínas do Citoesqueleto/metabolismo , Células HeLa , Mitose/fisiologia , Cadeias Leves de Miosina/metabolismo , Miosina Tipo II/metabolismo , Miosina Tipo II/fisiologia , Quinase de Cadeia Leve de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/fisiologia , Fosforilação , Quinases Associadas a rho/metabolismo
6.
Cytoskeleton (Hoboken) ; 77(9): 342-350, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32885903

RESUMO

The coordinated generation of mechanical forces by cardiac myocytes is required for proper heart function. Myofibrils are the functional contractile units of force production within individual cardiac myocytes. At the molecular level, myosin motors form cross-bridges with actin filaments and use ATP to convert chemical energy into mechanical forces. The energetic efficiency of the cross-bridge cycle is influenced by the viscous damping of myofibril contraction. The viscoelastic response of myofibrils is an emergent property of their individual mechanical components. Previous studies have implicated titin-actin interactions, cell-ECM adhesion, and microtubules as regulators of the viscoelastic response of myofibrils. Here we probed the viscoelastic response of myofibrils using laser-assisted dissection. As a proof-of-concept, we found actomyosin contractility was required to endow myofibrils with their viscoelastic response, with blebbistatin treatment resulting in decreased myofibril tension and viscous damping. Focal adhesion kinase (FAK) is a key regulator of cell-ECM adhesion, microtubule stability, and myofibril assembly. We found inhibition of FAK signaling altered the viscoelastic properties of myofibrils. Specifically, inhibition of FAK resulted in increased viscous damping of myofibril retraction following laser ablation. This damping was not associated with acute changes in the electrophysiological properties of cardiac myocytes. These results implicate FAK as a regulator of mechanical properties of myofibrils.


Assuntos
Adesões Focais/metabolismo , Miócitos Cardíacos/metabolismo , Miofibrilas/metabolismo , Humanos , Viscosidade
7.
iScience ; 23(4): 101015, 2020 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-32283523

RESUMO

MCL-1 is a well-characterized inhibitor of cell death that has also been shown to be a regulator of mitochondrial dynamics in human pluripotent stem cells. We used cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) to uncover whether MCL-1 is crucial for cardiac function and survival. Inhibition of MCL-1 by BH3 mimetics resulted in the disruption of mitochondrial morphology and dynamics as well as disorganization of the actin cytoskeleton. Interfering with MCL-1 function affects the homeostatic proximity of DRP-1 and MCL-1 at the outer mitochondrial membrane, resulting in decreased functionality of hiPSC-CMs. Cardiomyocytes display abnormal cardiac performance even after caspase inhibition, supporting a nonapoptotic activity of MCL-1 in hiPSC-CMs. BH3 mimetics targeting MCL-1 are promising anti-tumor therapeutics. Progression toward using BCL-2 family inhibitors, especially targeting MCL-1, depends on understanding its canonical function not only in preventing apoptosis but also in the maintenance of mitochondrial dynamics and function.

8.
Mol Biol Cell ; 31(12): 1273-1288, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32267210

RESUMO

Forces generated by heart muscle contraction must be balanced by adhesion to the extracellular matrix (ECM) and to other cells for proper heart function. Decades of data have suggested that cell-ECM adhesions are important for sarcomere assembly. However, the relationship between cell-ECM adhesions and sarcomeres assembling de novo remains untested. Sarcomeres arise from muscle stress fibers (MSFs) that are translocating on the top (dorsal) surface of cultured cardiomyocytes. Using an array of tools to modulate cell-ECM adhesion, we established a strong positive correlation between the extent of cell-ECM adhesion and sarcomere assembly. On the other hand, we found a strong negative correlation between the extent of cell-ECM adhesion and the rate of MSF translocation, a phenomenon also observed in nonmuscle cells. We further find a conserved network architecture that also exists in nonmuscle cells. Taken together, our results show that cell-ECM adhesions mediate coupling between the substrate and MSFs, allowing their maturation into sarcomere-containing myofibrils.


Assuntos
Miócitos Cardíacos/metabolismo , Miofibrilas/metabolismo , Fibras de Estresse/metabolismo , Actinas/metabolismo , Actinas/fisiologia , Técnicas de Cultura de Células/métodos , Junções Célula-Matriz/fisiologia , Matriz Extracelular/fisiologia , Humanos , Miócitos Cardíacos/fisiologia , Miofibrilas/fisiologia , Sarcômeros/fisiologia , Fibras de Estresse/fisiologia
9.
Cell Rep ; 31(1): 107477, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32268086

RESUMO

The mechanical properties of the actin cortex regulate shape changes during cell division, cell migration, and tissue morphogenesis. We show that modulation of myosin II (MII) filament composition allows tuning of surface tension at the cortex to maintain cell shape during cytokinesis. Our results reveal that MIIA generates cortex tension, while MIIB acts as a stabilizing motor and its inclusion in MII hetero-filaments reduces cortex tension. Tension generation by MIIA drives faster cleavage furrow ingression and bleb formation. We also show distinct roles for the motor and tail domains of MIIB in maintaining cytokinetic fidelity. Maintenance of cortical stability by the motor domain of MIIB safeguards against shape instability-induced chromosome missegregation, while its tail domain mediates cortical localization at the terminal stages of cytokinesis to mediate cell abscission. Because most non-muscle contractile systems are cortical, this tuning mechanism will likely be applicable to numerous processes driven by myosin-II contractility.


Assuntos
Forma Celular/fisiologia , Citocinese/fisiologia , Miosina Tipo II/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actinas/fisiologia , Animais , Células COS , Divisão Celular , Movimento Celular , Chlorocebus aethiops , Proteínas do Citoesqueleto/metabolismo , Células HeLa , Humanos , Morfogênese , Contração Muscular , Miosina Tipo II/fisiologia , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo
10.
Mol Biol Cell ; 30(9): 1051-1059, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30785846

RESUMO

Membrane blebs are specialized cellular protrusions that play diverse roles in processes such as cell division and cell migration. Blebbing can be divided into three distinct phases: bleb nucleation, bleb growth, and bleb retraction. Following nucleation and bleb growth, the actin cortex, comprising actin, cross-linking proteins, and nonmuscle myosin II (MII), begins to reassemble on the membrane. MII then drives the final phase, bleb retraction, which results in reintegration of the bleb into the cellular cortex. There are three MII paralogues with distinct biophysical properties expressed in mammalian cells: MIIA, MIIB, and MIIC. Here we show that MIIA specifically drives bleb retraction during cytokinesis. The motor domain and regulation of the nonhelical tailpiece of MIIA both contribute to its ability to drive bleb retraction. These experiments have also revealed a relationship between faster turnover of MIIA at the cortex and its ability to drive bleb retraction.


Assuntos
Vesícula/metabolismo , Miosina não Muscular Tipo IIA/fisiologia , Actinas/metabolismo , Animais , Células COS , Membrana Celular/metabolismo , Estruturas da Membrana Celular/metabolismo , Movimento Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Chlorocebus aethiops , Citocinese/fisiologia , Citoplasma/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células HeLa , Humanos , Miosina Tipo II/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo
11.
Curr Opin Cell Biol ; 56: 45-52, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30268802

RESUMO

The ability to divide is a fundamental property of a living cell. The 3D orientation of cell division is essential for embryogenesis, maintenance of tissue organization and architecture, as well as controlling cell fate. Much attention has been placed on the mitotic spindle's role in placing itself along the cell's longest axis, where a shape sensing mechanism between a population of microtubules extending from mitotic centrosomes to the cell cortex occurs. However, contractile forces at the cell cortex also likely play a decisive role in determining the final placement of daughter cells following division. In this review, we discuss recent literature that describes the role of these contractile forces and how these forces could be balanced by mitotic adhesion complexes.


Assuntos
Adesão Celular , Mitose , Fuso Acromático/metabolismo , Animais , Centrossomo/metabolismo , Humanos , Microtúbulos/metabolismo , Transdução de Sinais
12.
Elife ; 72018 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-30540249

RESUMO

The sarcomere is the contractile unit within cardiomyocytes driving heart muscle contraction. We sought to test the mechanisms regulating actin and myosin filament assembly during sarcomere formation. Therefore, we developed an assay using human cardiomyocytes to monitor sarcomere assembly. We report a population of muscle stress fibers, similar to actin arcs in non-muscle cells, which are essential sarcomere precursors. We show sarcomeric actin filaments arise directly from muscle stress fibers. This requires formins (e.g., FHOD3), non-muscle myosin IIA and non-muscle myosin IIB. Furthermore, we show short cardiac myosin II filaments grow to form ~1.5 µm long filaments that then 'stitch' together to form the stack of filaments at the core of the sarcomere (i.e., the A-band). A-band assembly is dependent on the proper organization of actin filaments and, as such, is also dependent on FHOD3 and myosin IIB. We use this experimental paradigm to present evidence for a unifying model of sarcomere assembly.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Miócitos Cardíacos/metabolismo , Sarcômeros/metabolismo , Fibras de Estresse/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Forminas , Células HeLa , Humanos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Microscopia Confocal , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/metabolismo , Fibras Musculares Esqueléticas/citologia , Miócitos Cardíacos/citologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIB/genética , Miosina não Muscular Tipo IIB/metabolismo , Interferência de RNA
13.
Sci Rep ; 6: 29846, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27432211

RESUMO

The geometry of the cleavage furrow during mitosis is often asymmetric in vivo and plays a critical role in stem cell differentiation and the relative positioning of daughter cells during development. Early observations of adhesive cell lines revealed asymmetry in the shape of the cleavage furrow, where the bottom (i.e., substrate attached side) of the cleavage furrow ingressed less than the top (i.e., unattached side). This data suggested substrate attachment could be regulating furrow ingression. Here we report a population of mitotic focal adhesions (FAs) controls the symmetry of the cleavage furrow. In single HeLa cells, stronger adhesion to the substrate directed less ingression from the bottom of the cell through a pathway including paxillin, focal adhesion kinase (FAK) and vinculin. Cell-cell contacts also direct ingression of the cleavage furrow in coordination with FAs in epithelial cells-MDCK-within monolayers and polarized cysts. In addition, mitotic FAs established 3D orientation of the mitotic spindle and the relative positioning of mother and daughter centrosomes. Therefore, our data reveals mitotic FAs as a key link between mitotic cell shape and spindle orientation, and may have important implications in our understanding stem cell homeostasis and tumorigenesis.


Assuntos
Forma Celular/genética , Adesões Focais/genética , Mitose/genética , Fuso Acromático/genética , Animais , Diferenciação Celular/genética , Centrossomo/metabolismo , Cães , Proteína-Tirosina Quinases de Adesão Focal/genética , Células HeLa , Humanos , Células Madin Darby de Rim Canino , Vinculina/genética
14.
Mol Biol Cell ; 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26960797

RESUMO

Cell movement and cytokinesis are facilitated by contractile forces generated by the molecular motor, non-muscle myosin II (NMII). NMII molecules form a filament (NMII-F) through interactions of their C-terminal rod domains, positioning groups of N-terminal motor domains on opposite sides. The NMII motors then bind and pull actin filaments toward the NMII-F, thus driving contraction. Inside of crawling cells, NMIIA-Fs form large macromolecular ensembles (i.e., NMIIA-F stacks) but how this occurs is unknown. Here we show NMIIA-F stacks are formed through two non-mutually exclusive mechanisms: expansion and concatenation. During expansion, NMIIA molecules within the NMIIA-F spread out concurrent with addition of new NMIIA molecules. Concatenation occurs when multiple NMIIA-F/NMIIA-F stacks move together and align. We found NMIIA-F stack formation was regulated by both motor-activity and the availability of surrounding actin filaments. Furthermore, our data showed expansion and concatenation also formed the contractile ring in dividing cells. Thus, interphase and mitotic cells share similar mechanisms for creating large contractile units, and these are likely to underlie how other myosin II-based contractile systems are assembled.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...