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












Base de datos
Intervalo de año de publicación
1.
Elife ; 122023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37921850

RESUMEN

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.


Asunto(s)
Miocitos Cardíacos , Sarcómeros , Sarcómeros/metabolismo , Miocitos Cardíacos/metabolismo , Actinina/metabolismo , Miofibrillas/metabolismo , Conectina/metabolismo , Programas Informáticos
3.
J Mol Cell Cardiol ; 179: 60-71, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37019277

RESUMEN

Standard transgenic cell line generation requires screening 100-1000s of colonies to isolate correctly edited cells. We describe CRISPRa On-Target Editing Retrieval (CRaTER) which enriches for cells with on-target knock-in of a cDNA-fluorescent reporter transgene by transient activation of the targeted locus followed by flow sorting to recover edited cells. We show CRaTER recovers rare cells with heterozygous, biallelic-editing of the transcriptionally-inactive MYH7 locus in human induced pluripotent stem cells (hiPSCs), enriching on average 25-fold compared to standard antibiotic selection. We leveraged CRaTER to enrich for heterozygous knock-in of a library of variants in MYH7, a gene in which missense mutations cause cardiomyopathies, and recovered hiPSCs with 113 different variants. We differentiated these hiPSCs to cardiomyocytes and show MHC-ß fusion proteins can localize as expected. Additionally, single-cell contractility analyses revealed cardiomyocytes with a pathogenic, hypertrophic cardiomyopathy-associated MYH7 variant exhibit salient HCM physiology relative to isogenic controls. Thus, CRaTER substantially reduces screening required for isolation of gene-edited cells, enabling generation of functional transgenic cell lines at unprecedented scale.


Asunto(s)
Cardiomiopatías , Cardiomiopatía Hipertrófica , Células Madre Pluripotentes Inducidas , Humanos , Edición Génica , Células Madre Pluripotentes Inducidas/metabolismo , Cardiomiopatías/metabolismo , Cardiomiopatía Hipertrófica/genética , Línea Celular , Mutación
4.
Cell Stem Cell ; 30(4): 396-414.e9, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-37028405

RESUMEN

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) offer a promising cell-based therapy for myocardial infarction. However, the presence of transitory ventricular arrhythmias, termed engraftment arrhythmias (EAs), hampers clinical applications. We hypothesized that EA results from pacemaker-like activity of hPSC-CMs associated with their developmental immaturity. We characterized ion channel expression patterns during maturation of transplanted hPSC-CMs and used pharmacology and genome editing to identify those responsible for automaticity in vitro. Multiple engineered cell lines were then transplanted in vivo into uninjured porcine hearts. Abolishing depolarization-associated genes HCN4, CACNA1H, and SLC8A1, along with overexpressing hyperpolarization-associated KCNJ2, creates hPSC-CMs that lack automaticity but contract when externally stimulated. When transplanted in vivo, these cells engrafted and coupled electromechanically with host cardiomyocytes without causing sustained EAs. This study supports the hypothesis that the immature electrophysiological prolife of hPSC-CMs mechanistically underlies EA. Thus, targeting automaticity should improve the safety profile of hPSC-CMs for cardiac remuscularization.


Asunto(s)
Edición Génica , Miocitos Cardíacos , Humanos , Animales , Porcinos , Miocitos Cardíacos/metabolismo , Línea Celular , Arritmias Cardíacas/genética , Arritmias Cardíacas/terapia , Arritmias Cardíacas/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos , Diferenciación Celular/genética
5.
bioRxiv ; 2023 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-36747685

RESUMEN

Standard transgenic cell line generation requires screening 100-1000s of colonies to isolate correctly edited cells. We describe CR ISPR a On- T arget E diting R etrieval (CRaTER) which enriches for cells with on-target knock-in of a cDNA-fluorescent reporter transgene by transient activation of the targeted locus followed by flow sorting to recover edited cells. We show CRaTER recovers rare cells with heterozygous, biallelic-editing of the transcriptionally-inactive MYH7 locus in human induced pluripotent stem cells (hiPSCs), enriching on average 25-fold compared to standard antibiotic selection. We leveraged CRaTER to enrich for heterozygous knock-in of a library of single nucleotide variants (SNVs) in MYH7 , a gene in which missense mutations cause cardiomyopathies, and recovered hiPSCs with 113 different MYH7 SNVs. We differentiated these hiPSCs to cardiomyocytes and show MYH7 fusion proteins can localize as expected. Thus, CRaTER substantially reduces screening required for isolation of gene-edited cells, enabling generation of transgenic cell lines at unprecedented scale.

6.
bioRxiv ; 2023 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-36711995

RESUMEN

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.

7.
Nat Commun ; 12(1): 6324, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732726

RESUMEN

Mutations in the cardiac splicing factor RBM20 lead to malignant dilated cardiomyopathy (DCM). To understand the mechanism of RBM20-associated DCM, we engineered isogenic iPSCs with DCM-associated missense mutations in RBM20 as well as RBM20 knockout (KO) iPSCs. iPSC-derived engineered heart tissues made from these cell lines recapitulate contractile dysfunction of RBM20-associated DCM and reveal greater dysfunction with missense mutations than KO. Analysis of RBM20 RNA binding by eCLIP reveals a gain-of-function preference of mutant RBM20 for 3' UTR sequences that are shared with amyotrophic lateral sclerosis (ALS) and processing-body associated RNA binding proteins (FUS, DDX6). Deep RNA sequencing reveals that the RBM20 R636S mutant has unique gene, splicing, polyadenylation and circular RNA defects that differ from RBM20 KO. Super-resolution microscopy verifies that mutant RBM20 maintains very limited nuclear localization potential; rather, the mutant protein associates with cytoplasmic processing bodies (DDX6) under basal conditions, and with stress granules (G3BP1) following acute stress. Taken together, our results highlight a pathogenic mechanism in cardiac disease through splicing-dependent and -independent pathways.


Asunto(s)
Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Mutación con Ganancia de Función , Mutación , Empalme del ARN , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/metabolismo , Cardiomiopatía Dilatada/genética , ARN Helicasas DEAD-box , ADN Helicasas , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Mutación Missense , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas Proto-Oncogénicas , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo
8.
Cell Rep ; 31(1): 107477, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32268086

RESUMEN

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.


Asunto(s)
Forma de la Célula/fisiología , Citocinesis/fisiología , Miosina Tipo II/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Actinas/fisiología , Animales , Células COS , División Celular , Movimiento Celular , Chlorocebus aethiops , Proteínas del Citoesqueleto/metabolismo , Células HeLa , Humanos , Morfogénesis , Contracción Muscular , Miosina Tipo II/fisiología , Miosina Tipo IIA no Muscular/metabolismo , Miosina Tipo IIB no Muscular/metabolismo
9.
Philos Trans R Soc Lond B Biol Sci ; 375(1792): 20190167, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31884915

RESUMEN

The phenomenon of ciliary coordination has garnered increasing attention in recent decades and multiple theories have been proposed to explain its occurrence in different biological systems. While hydrodynamic interactions are thought to dictate the large-scale coordinated activity of epithelial cilia for fluid transport, it is rather basal coupling that accounts for synchronous swimming gaits in model microeukaryotes such as Chlamydomonas. Unicellular ciliates present a fascinating yet understudied context in which coordination is found to persist in ciliary arrays positioned across millimetre scales on the same cell. Here, we focus on the ciliate Stentor coeruleus, chosen for its large size, complex ciliary organization, and capacity for cellular regeneration. These large protists exhibit ciliary differentiation between cortical rows of short body cilia used for swimming, and an anterior ring of longer, fused cilia called the membranellar band (MB). The oral cilia in the MB beat metachronously to produce strong feeding currents. Remarkably, upon injury, the MB can be shed and regenerated de novo. Here, we follow and track this developmental sequence in its entirety to elucidate the emergence of coordinated ciliary beating: from band formation, elongation, curling and final migration towards the cell anterior. We reveal a complex interplay between hydrodynamics and ciliary restructuring in Stentor, and highlight for the first time the importance of a ring-like topology for achieving long-range metachronism in ciliated structures. This article is part of the Theo Murphy meeting issue 'Unity and diversity of cilia in locomotion and transport'.


Asunto(s)
Cilios/fisiología , Cilióforos/fisiología , Regeneración , Cilióforos/crecimiento & desarrollo
10.
Cell ; 177(2): 428-445.e18, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30951670

RESUMEN

The heterogeneity of small extracellular vesicles and presence of non-vesicular extracellular matter have led to debate about contents and functional properties of exosomes. Here, we employ high-resolution density gradient fractionation and direct immunoaffinity capture to precisely characterize the RNA, DNA, and protein constituents of exosomes and other non-vesicle material. Extracellular RNA, RNA-binding proteins, and other cellular proteins are differentially expressed in exosomes and non-vesicle compartments. Argonaute 1-4, glycolytic enzymes, and cytoskeletal proteins were not detected in exosomes. We identify annexin A1 as a specific marker for microvesicles that are shed directly from the plasma membrane. We further show that small extracellular vesicles are not vehicles of active DNA release. Instead, we propose a new model for active secretion of extracellular DNA through an autophagy- and multivesicular-endosome-dependent but exosome-independent mechanism. This study demonstrates the need for a reassessment of exosome composition and offers a framework for a clearer understanding of extracellular vesicle heterogeneity.


Asunto(s)
Exosomas/metabolismo , Exosomas/fisiología , Anexina A1/metabolismo , Proteínas Argonautas/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , ADN/metabolismo , Exosomas/química , Vesículas Extracelulares , Femenino , Humanos , Lisosomas/metabolismo , Masculino , Proteínas/metabolismo , ARN/metabolismo
11.
J Cell Sci ; 132(7)2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30837285

RESUMEN

Basement membranes are an ancient form of animal extracellular matrix. As important structural and functional components of tissues, basement membranes are subject to environmental damage and must be repaired while maintaining functions. Little is known about how basement membranes get repaired. This paucity stems from a lack of suitable in vivo models for analyzing such repair. Here, we show that dextran sodium sulfate (DSS) directly damages the gut basement membrane when fed to adult Drosophila DSS becomes incorporated into the basement membrane, promoting its expansion while decreasing its stiffness, which causes morphological changes to the underlying muscles. Remarkably, two days after withdrawal of DSS, the basement membrane is repaired by all measures of analysis. We used this new damage model to determine that repair requires collagen crosslinking and replacement of damaged components. Genetic and biochemical evidence indicates that crosslinking is required to stabilize the newly incorporated repaired Collagen IV rather than to stabilize the damaged Collagen IV. These results suggest that basement membranes are surprisingly dynamic.


Asunto(s)
Membrana Basal/metabolismo , Colágeno Tipo IV/metabolismo , Matriz Extracelular/metabolismo , Laminina/metabolismo , Animales , Membrana Basal/efectos de los fármacos , Sulfato de Dextran , Drosophila melanogaster , Femenino , Masculino
12.
Curr Biol ; 29(1): 81-92.e5, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30581023

RESUMEN

Cell adhesion, morphogenesis, mechanosensing, and muscle contraction rely on contractile actomyosin bundles, where the force is produced through sliding of bipolar myosin II filaments along actin filaments. The assembly of contractile actomyosin bundles involves registered alignment of myosin II filaments and their subsequent fusion into large stacks. However, mechanisms underlying the assembly of myosin II stacks and their physiological functions have remained elusive. Here, we identified myosin-18B, an unconventional myosin, as a stable component of contractile stress fibers. Myosin-18B co-localized with myosin II motor domains in stress fibers and was enriched at the ends of myosin II stacks. Importantly, myosin-18B deletion resulted in drastic defects in the concatenation and persistent association of myosin II filaments with each other and thus led to severely impaired assembly of myosin II stacks. Consequently, lack of myosin-18B resulted in defective maturation of actomyosin bundles from their precursors in osteosarcoma cells. Moreover, myosin-18B knockout cells displayed abnormal morphogenesis, migration, and ability to exert forces to the environment. These results reveal a critical role for myosin-18B in myosin II stack assembly and provide evidence that myosin II stacks are important for a variety of vital processes in cells.


Asunto(s)
Contracción Muscular/fisiología , Miosina Tipo II/fisiología , Miosinas/metabolismo , Fibras de Estrés/fisiología , Proteínas Supresoras de Tumor/metabolismo , Actomiosina/metabolismo , Línea Celular Tumoral , Células HeLa , Humanos
13.
Elife ; 72018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30540249

RESUMEN

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.


Asunto(s)
Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/metabolismo , Sarcómeros/metabolismo , Fibras de Estrés/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Línea Celular , Línea Celular Tumoral , Forminas , Células HeLa , Humanos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Fibras Musculares Esqueléticas/citología , Miocitos Cardíacos/citología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo IIB no Muscular/genética , Miosina Tipo IIB no Muscular/metabolismo , Interferencia de ARN
14.
Cytoskeleton (Hoboken) ; 75(12): 545-549, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30126071

RESUMEN

How cellular contractile systems assemble has fascinated scientists for generations. The major molecule responsible for cellular force generation is the molecular motor, non-muscle myosin II (NMII). NMII molecules are organized into single myosin filaments and larger arrays of filaments called NMII stacks, which are capable of generating increasing amounts of force. The textbook model of NMII stack assembly is the Network Contraction Model, where ensembles of distinct NMII filaments condense into a NMII stack by pulling on actin filaments. While this model has been widely accepted for ~20 years, it has been difficult to test inside cells due to the small size of NMII filaments. Recently, interest in how NMII stacks form has been reinvigorated by the advent of super-resolution microscopy techniques which have afforded unprecedented resolution of NMII filaments inside cells. A number of recent publications using these techniques have called into question key aspects of the Network Contraction Model, and our understanding of how NMII stacks assemble.


Asunto(s)
Citoesqueleto de Actina , Modelos Biológicos , Miosina Tipo II , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Rastreo Celular , Humanos , Microscopía Electrónica , Miosina Tipo II/metabolismo , Miosina Tipo II/ultraestructura
15.
Sci Rep ; 8(1): 7546, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29765066

RESUMEN

Centromere-binding protein F (CENP-F) is a very large and complex protein with many and varied binding partners including components of the microtubule network. Numerous CENP-F functions impacting diverse cellular behaviors have been identified. Importantly, emerging data have shown that CENP-F loss- or gain-of-function has critical effects on human development and disease. Still, it must be noted that data at the single cardiac myocyte level examining the impact of CENP-F loss-of-function on fundamental cellular behavior is missing. To address this gap in our knowledge, we analyzed basic cell structure and function in cardiac myocytes devoid of CENP-F. We found many diverse structural abnormalities including disruption of the microtubule network impacting critical characteristics of the cardiac myocyte. This is the first report linking microtubule network malfunction to cardiomyopathy. Importantly, we also present data demonstrating a direct link between a CENP-F single nucleotide polymorphism (snp) and human cardiac disease. In a proximate sense, these data examining CENP-F function explain the cellular basis underlying heart disease in this genetic model and, in a larger sense, they will hopefully provide a platform upon which the field can explore diverse cellular outcomes in wide-ranging areas of research on this critical protein.


Asunto(s)
Cardiomiopatía Dilatada/genética , Proteínas Cromosómicas no Histona/genética , Insuficiencia Cardíaca/genética , Mutación con Pérdida de Función , Proteínas de Microfilamentos/genética , Miocitos Cardíacos/patología , Polimorfismo de Nucleótido Simple , Animales , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Proteínas Cromosómicas no Histona/metabolismo , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Insuficiencia Cardíaca/fisiopatología , Humanos , Uniones Intercelulares/patología , Ratones , Proteínas de Microfilamentos/metabolismo , Microtúbulos/patología , Miocitos Cardíacos/metabolismo , Volumen Sistólico
16.
J Cell Biol ; 216(12): 4053-4072, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29055011

RESUMEN

Contractile actomyosin bundles, stress fibers, are crucial for adhesion, morphogenesis, and mechanosensing in nonmuscle cells. However, the mechanisms by which nonmuscle myosin II (NM-II) is recruited to those structures and assembled into functional bipolar filaments have remained elusive. We report that UNC-45a is a dynamic component of actin stress fibers and functions as a myosin chaperone in vivo. UNC-45a knockout cells display severe defects in stress fiber assembly and consequent abnormalities in cell morphogenesis, polarity, and migration. Experiments combining structured-illumination microscopy, gradient centrifugation, and proteasome inhibition approaches revealed that a large fraction of NM-II and myosin-1c molecules fail to fold in the absence of UNC-45a. The remaining properly folded NM-II molecules display defects in forming functional bipolar filaments. The C-terminal UNC-45/Cro1/She4p domain of UNC-45a is critical for NM-II folding, whereas the N-terminal tetratricopeptide repeat domain contributes to the assembly of functional stress fibers. Thus, UNC-45a promotes generation of contractile actomyosin bundles through synchronized NM-II folding and filament-assembly activities.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Miosina Tipo II/metabolismo , Osteoblastos/metabolismo , Fibras de Estrés/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Polaridad Celular , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Miosina Tipo II/genética , Osteoblastos/ultraestructura , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Fibras de Estrés/ultraestructura , Repeticiones de Tetratricopéptidos
17.
ACS Appl Mater Interfaces ; 9(27): 22994-23006, 2017 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-28621931

RESUMEN

Human bone marrow derived mesenchymal stem cells (hMSCs) hold great promise for regenerative medicine due to their multipotent differentiation capacity and immunomodulatory capabilities. Substantial research has elucidated mechanisms by which extracellular cues regulate hMSC fate decisions, but considerably less work has addressed how material properties can be leveraged to maintain undifferentiated stem cells. Here, we show that synthetic culture substrates designed to exhibit moderate cell-repellency promote high stemness and low oxidative stress-two indicators of naïve, healthy stem cells-in commercial and patient-derived hMSCs. Furthermore, the material-mediated effect on cell behavior can be tuned by altering the molar percentage (mol %) and/or chain length of poly(ethylene glycol) (PEG), the repellant block linked to hydrophobic poly(ε-caprolactone) (PCL) in the copolymer backbone. Nano- and angstrom-scale characterization of the cell-material interface reveals that PEG interrupts the adhesive PCL domains in a chain-length-dependent manner; this prevents hMSCs from forming mature focal adhesions and subsequently promotes cell-cell adhesions that require connexin-43. This study is the first to demonstrate that intrinsic properties of synthetic materials can be tuned to regulate the stemness and redox capacity of hMSCs and provides new insight for designing highly scalable, programmable culture platforms for clinical translation.


Asunto(s)
Células Madre Mesenquimatosas , Diferenciación Celular , Humanos , Oxidación-Reducción , Polietilenglicoles , Medicina Regenerativa
18.
Sci Rep ; 6: 29846, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27432211

RESUMEN

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.


Asunto(s)
Forma de la Célula/genética , Adhesiones Focales/genética , Mitosis/genética , Huso Acromático/genética , Animales , Diferenciación Celular/genética , Centrosoma/metabolismo , Perros , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Vinculina/genética
19.
Mol Biol Cell ; 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26960797

RESUMEN

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.

20.
J Cell Biol ; 209(1): 11-2, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25869662

RESUMEN

A migrating cell must establish front-to-back polarity in order to move. In this issue, Juanes-Garcia et al. (2015. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201407059) report that a short serine-rich motif in nonmuscle myosin IIB is required to establish the cell's rear. This motif represents a new paradigm for what determines directional cell migration.


Asunto(s)
Polaridad Celular , Cadenas Pesadas de Miosina/fisiología , Miosina Tipo IIB no Muscular/fisiología , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...