Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(19): e2318413121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683993

RESUMEN

Determining the pathogenicity of hypertrophic cardiomyopathy-associated mutations in the ß-myosin heavy chain (MYH7) can be challenging due to its variable penetrance and clinical severity. This study investigates the early pathogenic effects of the incomplete-penetrant MYH7 G256E mutation on myosin function that may trigger pathogenic adaptations and hypertrophy. We hypothesized that the G256E mutation would alter myosin biomechanical function, leading to changes in cellular functions. We developed a collaborative pipeline to characterize myosin function across protein, myofibril, cell, and tissue levels to determine the multiscale effects on structure-function of the contractile apparatus and its implications for gene regulation and metabolic state. The G256E mutation disrupts the transducer region of the S1 head and reduces the fraction of myosin in the folded-back state by 33%, resulting in more myosin heads available for contraction. Myofibrils from gene-edited MYH7WT/G256E human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) exhibited greater and faster tension development. This hypercontractile phenotype persisted in single-cell hiPSC-CMs and engineered heart tissues. We demonstrated consistent hypercontractile myosin function as a primary consequence of the MYH7 G256E mutation across scales, highlighting the pathogenicity of this gene variant. Single-cell transcriptomic and metabolic profiling demonstrated upregulated mitochondrial genes and increased mitochondrial respiration, indicating early bioenergetic alterations. This work highlights the benefit of our multiscale platform to systematically evaluate the pathogenicity of gene variants at the protein and contractile organelle level and their early consequences on cellular and tissue function. We believe this platform can help elucidate the genotype-phenotype relationships underlying other genetic cardiovascular diseases.


Asunto(s)
Miosinas Cardíacas , Cardiomiopatía Hipertrófica , Células Madre Pluripotentes Inducidas , Contracción Miocárdica , Miocitos Cardíacos , Cadenas Pesadas de Miosina , Humanos , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Contracción Miocárdica/genética , Mutación , Mitocondrias/metabolismo , Mitocondrias/genética , Miofibrillas/metabolismo , Respiración de la Célula/genética
2.
bioRxiv ; 2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37333118

RESUMEN

Rationale: Over 200 mutations in the sarcomeric protein ß-myosin heavy chain (MYH7) have been linked to hypertrophic cardiomyopathy (HCM). However, different mutations in MYH7 lead to variable penetrance and clinical severity, and alter myosin function to varying degrees, making it difficult to determine genotype-phenotype relationships, especially when caused by rare gene variants such as the G256E mutation. Objective: This study aims to determine the effects of low penetrant MYH7 G256E mutation on myosin function. We hypothesize that the G256E mutation would alter myosin function, precipitating compensatory responses in cellular functions. Methods: We developed a collaborative pipeline to characterize myosin function at multiple scales (protein to myofibril to cell to tissue). We also used our previously published data on other mutations to compare the degree to which myosin function was altered. Results: At the protein level, the G256E mutation disrupts the transducer region of the S1 head and reduces the fraction of myosin in the folded-back state by 50.9%, suggesting more myosins available for contraction. Myofibrils isolated from hiPSC-CMs CRISPR-edited with G256E (MYH7 WT/G256E ) generated greater tension, had faster tension development and slower early phase relaxation, suggesting altered myosin-actin crossbridge cycling kinetics. This hypercontractile phenotype persisted in single-cell hiPSC-CMs and engineered heart tissues. Single-cell transcriptomic and metabolic profiling demonstrated upregulation of mitochondrial genes and increased mitochondrial respiration, suggesting altered bioenergetics as an early feature of HCM. Conclusions: MYH7 G256E mutation causes structural instability in the transducer region, leading to hypercontractility across scales, perhaps from increased myosin recruitment and altered crossbridge cycling. Hypercontractile function of the mutant myosin was accompanied by increased mitochondrial respiration, while cellular hypertrophy was modest in the physiological stiffness environment. We believe that this multi-scale platform will be useful to elucidate genotype-phenotype relationships underlying other genetic cardiovascular diseases.

3.
Proc Natl Acad Sci U S A ; 119(36): e2204688119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36037369

RESUMEN

Wnt signal transduction is controlled by the destruction complex (DC), a condensate comprising scaffold proteins and kinases that regulate ß-catenin stability. Overexpressed DC scaffolds undergo liquid-liquid phase separation (LLPS), but DC mesoscale organization at endogenous expression levels and its role in ß-catenin processing were previously unknown. Here, we find that DC LLPS is nucleated by the centrosome. Through a combination of CRISPR-engineered custom fluorescent tags, finite element simulations, and optogenetic tools that allow for manipulation of DC concentration and multivalency, we find that centrosomal nucleation drives processing of ß-catenin by colocalizing DC components to a single reaction crucible. Enriching GSK3ß partitioning on the centrosome controls ß-catenin processing and prevents Wnt-driven embryonic stem cell differentiation to mesoderm. Our findings demonstrate the role of nucleators in controlling biomolecular condensates and suggest tight integration between Wnt signal transduction and the cell cycle.


Asunto(s)
Centrosoma , Células Madre Embrionarias , Vía de Señalización Wnt , beta Catenina , Diferenciación Celular , Centrosoma/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Células Madre Embrionarias/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Mesodermo/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/genética , beta Catenina/metabolismo
4.
J Mol Cell Cardiol ; 168: 107-114, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35461823

RESUMEN

Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have advanced our ability to study the basic function of the heart and model cardiac diseases. Due to the complexities in stem cell culture and differentiation protocols, many researchers source their hiPSC-CMs from collaborators or commercial biobanks. Generally, the field has assumed the health of frozen cardiomyocytes is unchanged if the cells adhere to the substrate and commence beating. However, very few have investigated the effects of cryopreservation on hiPSC-CM's functional and transcriptional health at the cellular and molecular level. Here we review methods and challenges associated with cryopreservation, and examine the effects of cryopreservation on the functionality (contractility and calcium handling) and transcriptome of hiPSC-CMs from six healthy stem cell lines. Utilizing protein patterning methods to template physiological cell aspect ratios (7:1, length:width) in conjunction with polyacrylamide (PA) hydrogels, we measured changes in force generation and calcium handling of single hiPSC-CMs. We observed that cryopreservation altered the functionality and transcriptome of hiPSC-CMs towards larger sizes and contractile force as assessed by increased spread area and volume, single cell traction force microscopy and delayed calcium dynamics. hiPSC-CMs are broadly used for basic science research, regenerative medicine, and testing biological therapeutics. This study informs the design of experiments utilizing hiPSC-CMs to avoid confounding functional changes due to cryopreservation with other treatments.


Asunto(s)
Células Madre Pluripotentes Inducidas , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Criopreservación , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo
5.
Micromachines (Basel) ; 12(11)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34832798

RESUMEN

Human-induced pluripotent stem cell-derived cardiomyocytes are a potentially unlimited cell source and promising patient-specific in vitro model of cardiac diseases. Yet, these cells are limited by immaturity and population heterogeneity. Current in vitro studies aiming at better understanding of the mechanical and chemical cues in the microenvironment that drive cellular maturation involve deformable materials and precise manipulation of the microenvironment with, for example, micropatterns. Such microenvironment manipulation most often involves microfabrication protocols which are time-consuming, require cleanroom facilities and photolithography expertise. Here, we present a method to increase the scale of the fabrication pipeline, thereby enabling large-batch generation of shelf-stable microenvironment protein templates on glass chips. This decreases fabrication time and allows for more flexibility in the subsequent steps, for example, in tuning the material properties and the selection of extracellular matrix or cell proteins. Further, the fabrication of deformable hydrogels has been optimized for compatibility with these templates, in addition to the templates being able to be used to acquire protein patterns directly on the glass chips. With our approach, we have successfully controlled the shapes of cardiomyocytes seeded on Matrigel-patterned hydrogels.

6.
Materials (Basel) ; 14(8)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923751

RESUMEN

Recent advancements in tissue engineering and material science have radically improved in vitro culturing platforms to more accurately replicate human tissue. However, the transition to clinical relevance has been slow in part due to the lack of biologically compatible/relevant materials. In the present study, we marry the commonly used two-dimensional (2D) technique of electrospinning and a self-assembly process to construct easily reproducible, highly porous, three-dimensional (3D) nanofiber scaffolds for various tissue engineering applications. Specimens from biologically relevant polymers polycaprolactone (PCL) and gelatin were chemically cross-linked using the naturally occurring cross-linker genipin. Potential cytotoxic effects of the scaffolds were analyzed by culturing human dermal fibroblasts (HDF) up to 23 days. The 3D PCL/gelatin/genipin scaffolds produced here resemble the complex nanofibrous architecture found in naturally occurring extracellular matrix (ECM) and exhibit physiologically relevant mechanical properties as well as excellent cell cytocompatibility. Samples cross-linked with 0.5% genipin demonstrated the highest metabolic activity and proliferation rates for HDF. Scanning electron microscopy (SEM) images indicated excellent cell adhesion and the characteristic morphological features of fibroblasts in all tested samples. The three-dimensional (3D) PCL/gelatin/genipin scaffolds produced here show great potential for various 3D tissue-engineering applications such as ex vivo cell culturing platforms, wound healing, or tissue replacement.

7.
Sci Rep ; 11(1): 3026, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33542270

RESUMEN

Generating cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) has represented a significant advance in our ability to model cardiac disease. Current differentiation protocols, however, have limited use due to their production of heterogenous cell populations, primarily consisting of ventricular-like CMs. Here we describe the creation of two chamber-specific reporter hiPSC lines by site-directed genomic integration using CRISPR-Cas9 technology. In the MYL2-tdTomato reporter, the red fluorescent tdTomato was inserted upstream of the 3' untranslated region of the Myosin Light Chain 2 (MYL2) gene in order faithfully label hiPSC-derived ventricular-like CMs while avoiding disruption of endogenous gene expression. Similarly, in the SLN-CFP reporter, Cyan Fluorescent Protein (CFP) was integrated downstream of the coding region of the atrial-specific gene, Sarcolipin (SLN). Purification of tdTomato+ and CFP+ CMs using flow cytometry coupled with transcriptional and functional characterization validated these genetic tools for their use in the isolation of bona fide ventricular-like and atrial-like CMs, respectively. Finally, we successfully generated a double reporter system allowing for the isolation of both ventricular and atrial CM subtypes within a single hiPSC line. These tools provide a platform for chamber-specific hiPSC-derived CM purification and analysis in the context of atrial- or ventricular-specific disease and therapeutic opportunities.


Asunto(s)
Diferenciación Celular/genética , Atrios Cardíacos/crecimiento & desarrollo , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Sistemas CRISPR-Cas/genética , Miosinas Cardíacas/genética , Proteínas Fluorescentes Verdes , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/patología , Miocitos Cardíacos/citología , Cadenas Ligeras de Miosina/genética
8.
Cell Stem Cell ; 27(1): 50-63.e5, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32619518

RESUMEN

Modulating signaling pathways including Wnt and Hippo can induce cardiomyocyte proliferation in vivo. Applying these signaling modulators to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro can expand CMs modestly (<5-fold). Here, we demonstrate massive expansion of hiPSC-CMs in vitro (i.e., 100- to 250-fold) by glycogen synthase kinase-3ß (GSK-3ß) inhibition using CHIR99021 and concurrent removal of cell-cell contact. We show that GSK-3ß inhibition suppresses CM maturation, while contact removal prevents CMs from cell cycle exit. Remarkably, contact removal enabled 10 to 25 times greater expansion beyond GSK-3ß inhibition alone. Mechanistically, persistent CM proliferation required both LEF/TCF activity and AKT phosphorylation but was independent from yes-associated protein (YAP) signaling. Engineered heart tissues from expanded hiPSC-CMs showed comparable contractility to those from unexpanded hiPSC-CMs, demonstrating uncompromised cellular functionality after expansion. In summary, we uncovered a molecular interplay that enables massive hiPSC-CM expansion for large-scale drug screening and tissue engineering applications.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3 beta , Humanos , Miocitos Cardíacos
9.
Circ Res ; 125(4): 379-397, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31284824

RESUMEN

RATIONALE: The cardiac conduction system (CCS) consists of distinct components including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers. Despite an essential role for the CCS in heart development and function, the CCS has remained challenging to interrogate because of inherent obstacles including small cell numbers, large cell-type heterogeneity, complex anatomy, and difficulty in isolation. Single-cell RNA-sequencing allows for genome-wide analysis of gene expression at single-cell resolution. OBJECTIVE: Assess the transcriptional landscape of the entire CCS at single-cell resolution by single-cell RNA-sequencing within the developing mouse heart. METHODS AND RESULTS: Wild-type, embryonic day 16.5 mouse hearts (n=6 per zone) were harvested and 3 zones of microdissection were isolated, including: Zone I-sinoatrial node region; Zone II-atrioventricular node/His region; and Zone III-bundle branch/Purkinje fiber region. Tissue was digested into single-cell suspensions, cells isolated, mRNA reverse transcribed, and barcoded before high-throughput sequencing and bioinformatics analyses. Single-cell RNA-sequencing was performed on over 22 000 cells, and all major cell types of the murine heart were successfully captured including bona fide clusters of cells consistent with each major component of the CCS. Unsupervised weighted gene coexpression network analysis led to the discovery of a host of novel CCS genes, a subset of which were validated using fluorescent in situ hybridization as well as whole-mount immunolabeling with volume imaging (iDISCO+) in 3 dimensions on intact mouse hearts. Further, subcluster analysis unveiled isolation of distinct CCS cell subtypes, including the clinically relevant but poorly characterized transitional cells that bridge the CCS and surrounding myocardium. CONCLUSIONS: Our study represents the first comprehensive assessment of the transcriptional profiles from the entire CCS at single-cell resolution and provides a characterization in the context of development and disease.


Asunto(s)
Sistema de Conducción Cardíaco/metabolismo , Transcriptoma , Animales , Sistema de Conducción Cardíaco/citología , Sistema de Conducción Cardíaco/embriología , Ratones , RNA-Seq , Análisis de la Célula Individual
10.
Sci Rep ; 8(1): 6618, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29700394

RESUMEN

Bioactive lipids such as sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) regulate diverse processes including cell proliferation, differentiation, and migration. However, their roles in cardiac differentiation and cardiomyocyte proliferation have not been explored. Using a 96-well differentiation platform for generating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) we found that S1P and LPA can independently enhance cardiomyocyte generation when administered at an early stage of differentiation. We showed that the combined S1P and LPA treatment of undifferentiated hiPSCs resulted in increased nuclear accumulation of ß-catenin, the canonical Wnt signaling pathway mediator, and synergized with CHIR99021, a glycogen synthase kinase 3 beta inhibitor, to enhance mesodermal induction and subsequent cardiac differentiation. At later stages of cardiac differentiation, the addition of S1P and LPA resulted in cell cycle initiation in hiPSC-CMs, an effect mediated through increased ERK signaling. Although the addition of S1P and LPA alone was insufficient to induce cell division, it was able to enhance ß-catenin-mediated hiPSC-CM proliferation. In summary, we demonstrated a developmental stage-specific effect of bioactive lipids to enhance hiPSC-CM differentiation and proliferation via modulating the effect of canonical Wnt/ß-catenin and ERK signaling. These findings may improve hiPSC-CM generation for cardiac disease modeling, precision medicine, and regenerative therapies.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Lípidos/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Biomarcadores , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mesodermo/citología , Mesodermo/efectos de los fármacos , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
11.
Sci Rep ; 7(1): 12590, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974782

RESUMEN

During normal lifespan, the mammalian heart undergoes limited renewal of cardiomyocytes. While the exact mechanism for this renewal remains unclear, two possibilities have been proposed: differentiated myocyte replication and progenitor/immature cell differentiation. This study aimed to characterize a population of cardiomyocyte precursors in the neonatal heart and to determine their requirement for cardiac development. By tracking the expression of an embryonic Nkx2.5 cardiac enhancer, we identified cardiomyoblasts capable of differentiation into striated cardiomyocytes in vitro. Genome-wide expression profile of neonatal Nkx2.5+ cardiomyoblasts showed the absence of sarcomeric gene and the presence of cardiac transcription factors. To determine the lineage contribution of the Nkx2.5+ cardiomyoblasts, we generated a doxycycline suppressible Cre transgenic mouse under the regulation of the Nkx2.5 enhancer and showed that neonatal Nkx2.5+ cardiomyoblasts mature into cardiomyocytes in vivo. Ablation of neonatal cardiomyoblasts resulted in ventricular hypertrophy and dilation, supporting a functional requirement of the Nkx2.5+ cardiomyoblasts. This study provides direct lineage tracing evidence that a cardiomyoblast population contributes to cardiogenesis in the neonatal heart. The cell population identified here may serve as a promising therapeutic for pediatric cardiac regeneration.


Asunto(s)
Cardiomegalia/genética , Linaje de la Célula/genética , Proteína Homeótica Nkx-2.5/genética , Desarrollo de Músculos/genética , Animales , Animales Recién Nacidos , Cardiomegalia/patología , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Ratones Transgénicos , Mioblastos Cardíacos/metabolismo , Mioblastos Cardíacos/patología , Miocitos Cardíacos/metabolismo
12.
Biomaterials ; 131: 111-120, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28384492

RESUMEN

Engineering 3D human cardiac tissues is of great importance for therapeutic and pharmaceutical applications. As cardiac tissue substitutes, extracellular matrix-derived hydrogels have been widely explored. However, they exhibit premature degradation and their stiffness is often orders of magnitude lower than that of native cardiac tissue. There are no reports on establishing interconnected cardiomyocytes in 3D hydrogels at physiologically-relevant cell density and matrix stiffness. Here we bioengineer human cardiac microtissues by encapsulating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in chemically-crosslinked gelatin hydrogels (1.25 × 108/mL) with tunable stiffness and degradation. In comparison to the cells in high stiffness (16 kPa)/slow degrading hydrogels, hiPSC-CMs in low stiffness (2 kPa)/fast degrading and intermediate stiffness (9 kPa)/intermediate degrading hydrogels exhibit increased intercellular network formation, α-actinin and connexin-43 expression, and contraction velocity. Only the 9 kPa microtissues exhibit organized sarcomeric structure and significantly increased contractile stress. This demonstrates that muscle-mimicking stiffness together with robust cellular interconnection contributes to enhancement in sarcomeric organization and contractile function of the engineered cardiac tissue. This study highlights the importance of intercellular connectivity, physiologically-relevant cell density, and matrix stiffness to best support 3D cardiac tissue engineering.


Asunto(s)
Materiales Biocompatibles/química , Gelatina/química , Hidrogeles/química , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Ingeniería de Tejidos/métodos , Fenómenos Biomecánicos , Biomimética/métodos , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Células Inmovilizadas/química , Reactivos de Enlaces Cruzados/química , Elasticidad , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA