Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(7): 1796-1810.e20, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30528432

RESUMEN

The 9p21.3 cardiovascular disease locus is the most influential common genetic risk factor for coronary artery disease (CAD), accounting for ∼10%-15% of disease in non-African populations. The ∼60 kb risk haplotype is human-specific and lacks coding genes, hindering efforts to decipher its function. Here, we produce induced pluripotent stem cells (iPSCs) from risk and non-risk individuals, delete each haplotype using genome editing, and generate vascular smooth muscle cells (VSMCs). Risk VSMCs exhibit globally altered transcriptional networks that intersect with previously identified CAD risk genes and pathways, concomitant with aberrant adhesion, contraction, and proliferation. Unexpectedly, deleting the risk haplotype rescues VSMC stability, while expressing the 9p21.3-associated long non-coding RNA ANRIL induces risk phenotypes in non-risk VSMCs. This study shows that the risk haplotype selectively predisposes VSMCs to adopt a cell state associated with CAD phenotypes, defines new VSMC-based networks of CAD risk genes, and establishes haplotype-edited iPSCs as powerful tools for functionally annotating the human genome.


Asunto(s)
Cromosomas Humanos Par 9 , Enfermedad de la Arteria Coronaria , Edición Génica , Haplotipos , Células Madre Pluripotentes Inducidas , Polimorfismo de Nucleótido Simple , Anciano , Anciano de 80 o más Años , Cromosomas Humanos Par 9/genética , Cromosomas Humanos Par 9/metabolismo , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Femenino , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transcripción Genética
2.
Proc Natl Acad Sci U S A ; 120(24): e2217122120, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37276403

RESUMEN

9p21.3 locus polymorphisms have the strongest correlation with coronary artery disease, but as a noncoding locus, disease connection is enigmatic. The lncRNA ANRIL found in 9p21.3 may regulate vascular smooth muscle cell (VSMC) phenotype to contribute to disease risk. We observed significant heterogeneity in induced pluripotent stem cell-derived VSMCs from patients homozygous for risk versus isogenic knockout or nonrisk haplotypes. Subpopulations of risk haplotype cells exhibited variable morphology, proliferation, contraction, and adhesion. When sorted by adhesion, risk VSMCs parsed into synthetic and contractile subpopulations, i.e., weakly adherent and strongly adherent, respectively. Of note, >90% of differentially expressed genes coregulated by haplotype and adhesion and were associated with Rho GTPases, i.e., contractility. Weakly adherent subpopulations expressed more short isoforms of ANRIL, and when overexpressed in knockout cells, ANRIL suppressed adhesion, contractility, and αSMA expression. These data suggest that variable lncRNA penetrance may drive mixed functional outcomes that confound pathology.


Asunto(s)
Enfermedad de la Arteria Coronaria , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Músculo Liso Vascular/metabolismo , Plasticidad de la Célula/genética , Enfermedad de la Arteria Coronaria/genética , Fenotipo , Miocitos del Músculo Liso/metabolismo , Proliferación Celular , Células Cultivadas
3.
J Cell Sci ; 136(23)2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37994565

RESUMEN

Matrix remodeling outcomes largely dictate patient survival post myocardial infarction. Moreover, human-restricted noncoding regulatory elements have been shown to worsen fibrosis, but their mechanism of action remains elusive. Here, we demonstrate, using induced pluripotent stem cell-derived cardiac fibroblasts (iCFs), that inflammatory ligands abundant in the remodeling heart after infarction activate AP-1 transcription factor signaling pathways resulting in fibrotic responses. This observed signaling induces deposition of fibronectin matrix and is further capable of supporting immune cell adhesion; pathway inhibition blocks iCF matrix production and cell adhesion. Polymorphisms in the noncoding regulatory elements within the 9p21 locus (also referred to as ANRIL) redirect stress programs, and in iCFs, they transcriptionally silence the AP-1 inducible transcription factor GATA5. The presence of these polymorphisms modulate iCF matrix production and assembly and reduce cell-cell signaling. These data suggest that this signaling axis is a critical modulator of cardiac disease models and might be influenced by noncoding regulatory elements.


Asunto(s)
Miocardio , Factor de Transcripción AP-1 , Humanos , Fibroblastos/metabolismo , Fibrosis , Corazón , Miocardio/metabolismo , Transducción de Señal , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
4.
Am J Physiol Cell Physiol ; 326(4): C1193-C1202, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38581669

RESUMEN

Satellite cells (SCs) and fibroadipogenic progenitors (FAPs) are progenitor populations found in muscle that form new myofibers postinjury. Muscle development, regeneration, and tissue-engineering experiments require robust progenitor populations, yet their isolation and expansion are difficult given their scarcity in muscle, limited muscle biopsy sizes in humans, and lack of methodological detail in the literature. Here, we investigated whether a dispase and collagenase type 1 and 2 cocktail could allow dual isolation of SCs and FAPs, enabling significantly increased yield from human skeletal muscle. Postdissociation, we found that single cells could be sorted into CD56 + CD31-CD45- (SC) and CD56-CD31-CD45- (FAP) cell populations, expanded in culture, and characterized for lineage-specific marker expression and differentiation capacity; we obtained ∼10% SCs and ∼40% FAPs, with yields twofold better than what is reported in current literature. SCs were PAX7+ and retained CD56 expression and myogenic fusion potential after multiple passages, expanding up to 1012 cells. Conversely, FAPs expressed CD140a and differentiated into either fibroblasts or adipocytes upon induction. This study demonstrates robust isolation of both SCs and FAPs from the same muscle sample with SC recovery more than two times higher than previously reported, which could enable translational studies for muscle injuries.NEW & NOTEWORTHY We demonstrated that a dispase/collagenase cocktail allows for simultaneous isolation of SCs and FAPs with 2× higher SC yield compared with other studies. We provide a thorough characterization of SC and FAP in vitro expansion that other studies have not reported. Following our dissociation, SCs and FAPs were able to expand by up to 1012 cells before reaching senescence and maintained differentiation capacity in vitro demonstrating their efficacy for clinical translation for muscle injury.


Asunto(s)
Endopeptidasas , Músculo Esquelético , Células Satélite del Músculo Esquelético , Humanos , Músculo Esquelético/metabolismo , Diferenciación Celular/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Fibroblastos/metabolismo
5.
Nature ; 560(7720): 655-660, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30135582

RESUMEN

Mammalian cells are surrounded by neighbouring cells and extracellular matrix (ECM), which provide cells with structural support and mechanical cues that influence diverse biological processes1. The Hippo pathway effectors YAP (also known as YAP1) and TAZ (also known as WWTR1) are regulated by mechanical cues and mediate cellular responses to ECM stiffness2,3. Here we identified the Ras-related GTPase RAP2 as a key intracellular signal transducer that relays ECM rigidity signals to control mechanosensitive cellular activities through YAP and TAZ. RAP2 is activated by low ECM stiffness, and deletion of RAP2 blocks the regulation of YAP and TAZ by stiffness signals and promotes aberrant cell growth. Mechanistically, matrix stiffness acts through phospholipase Cγ1 (PLCγ1) to influence levels of phosphatidylinositol 4,5-bisphosphate and phosphatidic acid, which activates RAP2 through PDZGEF1 and PDZGEF2 (also known as RAPGEF2 and RAPGEF6). At low stiffness, active RAP2 binds to and stimulates MAP4K4, MAP4K6, MAP4K7 and ARHGAP29, resulting in activation of LATS1 and LATS2 and inhibition of YAP and TAZ. RAP2, YAP and TAZ have pivotal roles in mechanoregulated transcription, as deletion of YAP and TAZ abolishes the ECM stiffness-responsive transcriptome. Our findings show that RAP2 is a molecular switch in mechanotransduction, thereby defining a mechanosignalling pathway from ECM stiffness to the nucleus.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Transformación Celular Neoplásica , Matriz Extracelular/química , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Femenino , Proteínas Activadoras de GTPasa/metabolismo , Quinasas del Centro Germinal , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HEK293 , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Proteínas del Tejido Nervioso/metabolismo , Fosfolipasa C gamma/metabolismo , Fosfoproteínas/metabolismo , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Transcriptoma , Proteínas Señalizadoras YAP , Proteínas de Unión al GTP rap/genética
6.
J Mol Cell Cardiol ; 164: 58-68, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34826415

RESUMEN

Since the initial isolation of human embryonic stem cells and subsequent discovery of reprogramming methods for somatic cells, thousands of protocols have been developed to create each of the hundreds of cell types found in-vivo with significant focus on disease-prone systems, e.g., cardiovascular. Robust protocols exist for many of these cell types, except for cardiac fibroblasts (CF). Very recently, several competing methods have been developed to generate these cells through a developmentally conserved epicardial pathway. Such methods generate epicardial cells, but here we report that prolonged exposure to growth factors such as bFGF induces fibroblast spindle-like morphology and similar chromatin architecture to primary CFs. Media conditions for growth and assays are provided, as well as suggestions for seeding densities and timepoints for protein harvest of extracellular matrix. We demonstrate marker expression and matrix competency of resultant cells as shown next to primary human cardiac fibroblasts. These methods provide additional guidance to the original protocol and result in an increasingly stable phenotype.


Asunto(s)
Células Madre Embrionarias Humanas , Células Madre Pluripotentes Inducidas , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Corazón , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
7.
J Cell Sci ; 133(24)2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33243756

RESUMEN

A lack of biological markers has limited our ability to identify the invasive cells responsible for glioblastoma multiforme (GBM). To become migratory and invasive, cells must downregulate matrix adhesions, which could be a physical marker of invasive potential. We engineered murine astrocytes with common GBM mutations, e.g. Ink4a (Ink) or PTEN deletion and expressing a constitutively active EGF receptor truncation (EGFRvIII), to elucidate their effect on adhesion. While loss of Ink or PTEN did not affect adhesion, counterparts expressing EGFRvIII were significantly less adhesive. EGFRvIII reduced focal adhesion size and number, and these cells - with more labile adhesions - displayed enhanced migration. Regulation appears to depend not on physical receptor association to integrins but, rather, on the activity of the receptor kinase, resulting in transcriptional integrin repression. Interestingly, EGFRvIII intrinsic signals can be propagated by cytokine crosstalk to cells expressing wild-type EGFR, resulting in reduced adhesion and enhanced migration. These data identify potential intrinsic and extrinsic mechanisms that gliomas use to invade surrounding parenchyma.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/genética , Glioma/genética , Ratones , Transducción de Señal
8.
Proc Natl Acad Sci U S A ; 116(9): 3502-3507, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30755531

RESUMEN

Breast cancer development is associated with increasing tissue stiffness over years. To more accurately mimic the onset of gradual matrix stiffening, which is not feasible with conventional static hydrogels, mammary epithelial cells (MECs) were cultured on methacrylated hyaluronic acid hydrogels whose stiffness can be dynamically modulated from "normal" (<150 Pascals) to "malignant" (>3,000 Pascals) via two-stage polymerization. MECs form and remain as spheroids, but begin to lose epithelial characteristics and gain mesenchymal morphology upon matrix stiffening. However, both the degree of matrix stiffening and culture time before stiffening play important roles in regulating this conversion as, in both cases, a subset of mammary spheroids remained insensitive to local matrix stiffness. This conversion depended neither on colony size nor cell density, and MECs did not exhibit "memory" of prior niche when serially cultured through cycles of compliant and stiff matrices. Instead, the transcription factor Twist1, transforming growth factor ß (TGFß), and YAP activation appeared to modulate stiffness-mediated signaling; when stiffness-mediated signals were blocked, collective MEC phenotypes were reduced in favor of single MECs migrating away from spheroids. These data indicate a more complex interplay of time-dependent stiffness signaling, spheroid structure, and soluble cues that regulates MEC plasticity than suggested by previous models.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/genética , Mecanotransducción Celular/genética , Fosfoproteínas/genética , Factor de Crecimiento Transformador beta/genética , Proteína 1 Relacionada con Twist/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Hidrogeles/química , Comunicación Paracrina/genética , Transducción de Señal/genética , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Factores de Transcripción , Proteínas Señalizadoras YAP
9.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055055

RESUMEN

Dilated cardiomyopathy (DCM) is a life-threatening form of heart disease that is typically characterized by progressive thinning of the ventricular walls, chamber dilation, and systolic dysfunction. Multiple mutations in the gene encoding filamin C (FLNC), an actin-binding cytoskeletal protein in cardiomyocytes, have been found in patients with DCM. However, the mechanisms that lead to contractile impairment and DCM in patients with FLNC variants are poorly understood. To determine how FLNC regulates systolic force transmission and DCM remodeling, we used an inducible, cardiac-specific FLNC-knockout (icKO) model to produce a rapid onset of DCM in adult mice. Loss of FLNC reduced systolic force development in single cardiomyocytes and isolated papillary muscles but did not affect twitch kinetics or calcium transients. Electron and immunofluorescence microscopy showed significant defects in Z-disk alignment in icKO mice and altered myofilament lattice geometry. Moreover, a loss of FLNC induces a softening myocyte cortex and structural adaptations at the subcellular level that contribute to disrupted longitudinal force production during contraction. Spatially explicit computational models showed that these structural defects could be explained by a loss of inter-myofibril elastic coupling at the Z-disk. Our work identifies FLNC as a key regulator of the multiscale ultrastructure of cardiomyocytes and therefore plays an important role in maintaining systolic mechanotransmission pathways, the dysfunction of which may be key in driving progressive DCM.


Asunto(s)
Biomarcadores , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/metabolismo , Filaminas/deficiencia , Predisposición Genética a la Enfermedad , Miocitos Cardíacos/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Cardiomiopatía Dilatada/diagnóstico , Costameras/genética , Costameras/metabolismo , Modelos Animales de Enfermedad , Femenino , Filaminas/metabolismo , Expresión Génica , Estudios de Asociación Genética , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación , Contracción Miocárdica/genética
10.
J Cell Sci ; 132(1)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30559248

RESUMEN

Tumors are composed of heterogeneous phenotypes, each having different sensitivities to the microenvironment. One microenvironment characteristic - matrix stiffness - helps to regulate malignant transformation and invasion in mammary tumors, but its influence on oral squamous cell carcinoma (OSCC) is unclear. We observed that, on stiff matrices, a highly invasive OSCC cell line (SCC25) comprising a low E-cad to N-cad ratio (InvH/E:NL; SCC25) had increased migration velocity and decreased adhesion strength compared to a less invasive OSCC cell line (Cal27) with high E-cad to N-cad ratio (InvL/E:NH; Cal27). However, InvL/E:NH cells acquire a mesenchymal signature and begin to migrate faster when exposed to prolonged time on a stiff niche, suggesting that cells can be mechanically conditioned. Owing to increased focal adhesion assembly, InvL/E:NH cells migrated faster, which could be reduced when increasing integrin affinity with high divalent cation concentrations. Mirroring these data in human patients, we observed that collagen organization, an indicator of matrix stiffness, was increased with advanced disease and correlated with early recurrence. Consistent with epithelial tumors, our data suggest that OSCC cells are mechanically sensitive and that their contribution to tumor progression is mediated in part by this sensitivity.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Carcinoma de Células Escamosas/patología , Movimiento Celular , Transformación Celular Neoplásica/patología , Colágeno/metabolismo , Transición Epitelial-Mesenquimal , Matriz Extracelular/patología , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas/metabolismo , Adhesión Celular , Transformación Celular Neoplásica/metabolismo , Matriz Extracelular/metabolismo , Humanos , Neoplasias de la Boca/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral
11.
Am J Physiol Heart Circ Physiol ; 320(6): H2211-H2221, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33769920

RESUMEN

Aside from the first week postnatal, murine heart regeneration is restricted and responses to damage follow classic fibrotic remodeling. Recent transcriptomic analyses have suggested that significant cross talk with the sterile immune response could maintain a more embryonic-like signaling network that promotes acute, transient responses. However, with age, this response-likely mediated by neonatal yolk sac macrophages-then transitions to classical macrophage-mediated, cardiac fibroblast (CF)-based remodeling of the extracellular matrix (ECM) after myocardial infarction (MI). The molecular mechanisms that govern the change with age and drive fibrosis via inflammation are poorly understood. Using multiple ribonucleic acid sequencing (RNA-Seq) datasets, we attempt to resolve the relative contributions of CFs and macrophages in the bulk-healing response of regenerative (postnatal day 1) and nonregenerative hearts (postnatal day 8+). We performed an analysis of bulk RNA-Seq datasets from myocardium and cardiac fibroblasts as well as a single-cell RNA-Seq dataset from cardiac macrophages. MI-specific pathway differences revealed that nonregenerative hearts generated more ECM and had larger matricellular responses correlating with inflammation, produced greater chemotactic gradients to recruit macrophages, and expressed receptors for danger-associated molecular patterns at higher levels than neonates. These changes could result in elevated stress-response pathways compared with neonates, converging at NF-κB and activator protein-1 (AP-1) signaling. Profibrotic gene programs, which greatly diverge on day 3 post MI, lay the foundation for chronic fibrosis, and thus postnatal hearts older than 7 days typically exhibit significantly less regeneration. Our analyses suggest that the macrophage ontogenetic shift in the heart postnatally could result in detrimental stress signaling that suppresses regeneration.NEW & NOTEWORTHY Immediately postnatal mammalian hearts are able to regenerate after infarction, but the cells, pathways, and molecules that regulate this behavior are unclear. By comparing RNA-Seq datasets from regenerative mouse hearts and older, nonregenerative hearts, we are able to identify biological processes that are hallmarks of regeneration. We find that sterile inflammatory processes are upregulated in nonregenerative hearts, initiating profibrotic gene programs 3 days after myocardial infarction that can cause myocardial disease.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis/genética , Inflamación/genética , Macrófagos/metabolismo , Infarto del Miocardio/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Regeneración/genética , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibrosis/metabolismo , Fibrosis/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Corazón/fisiología , Inflamación/metabolismo , Inflamación/patología , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , RNA-Seq , Regeneración/fisiología , Análisis de la Célula Individual
12.
Development ; 144(23): 4261-4270, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29183939

RESUMEN

Stem cells interpret signals from their microenvironment while simultaneously modifying the niche through secreting factors and exerting mechanical forces. Many soluble stem cell cues have been determined over the past century, but in the past decade, our molecular understanding of mechanobiology has advanced to explain how passive and active forces induce similar signaling cascades that drive self-renewal, migration, differentiation or a combination of these outcomes. Improvements in stem cell culture methods, materials and biophysical tools that assess function have improved our understanding of these cascades. Here, we summarize these advances and offer perspective on ongoing challenges.


Asunto(s)
Células Madre/fisiología , Actomiosina/fisiología , Animales , Fenómenos Biomecánicos , Fenómenos Biofísicos , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Matriz Extracelular/fisiología , Humanos , Transducción de Señal , Nicho de Células Madre/fisiología , Células Madre/citología , Factores de Transcripción/metabolismo
13.
Exp Cell Res ; 377(1-2): 103-108, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30794804

RESUMEN

Tissues are continuously exposed to forces in vivo, whether from fluid pressure in an artery from our blood or compressive forces on joints from our body weight. The forces that cells are exposed to arise almost immediately after conception; it is therefore important to understand how forces shape stem cell differentiation into lineage committed cells, how they help organize cells into tissues, and how forces can cause or exacerbate disease. No tissue is exempt, but cardiovascular tissues in particular are exposed to these forces. While animal models have been used extensively in the past, there is growing recognition of their limitations when modeling disease complexity or human genetics. In this mini review, we summarize current understanding of the mechanical influences on the differentiation of cardiovascular progeny, how the transduction of forces influence the onset of disease, and how engineering approaches applied to this problem have yielded systems that create mature-like human tissues in vitro in which to assess the impact of disease on cell function.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Sistema Cardiovascular/citología , Diferenciación Celular , Mecanotransducción Celular , Células Madre/citología , Estrés Mecánico , Animales , Humanos
14.
Proc Natl Acad Sci U S A ; 114(22): 5647-5652, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28507138

RESUMEN

The spatial presentation of mechanical information is a key parameter for cell behavior. We have developed a method of polymerization control in which the differential diffusion distance of unreacted cross-linker and monomer into a prepolymerized hydrogel sink results in a tunable stiffness gradient at the cell-matrix interface. This simple, low-cost, robust method was used to produce polyacrylamide hydrogels with stiffness gradients of 0.5, 1.7, 2.9, 4.5, 6.8, and 8.2 kPa/mm, spanning the in vivo physiological and pathological mechanical landscape. Importantly, three of these gradients were found to be nondurotactic for human adipose-derived stem cells (hASCs), allowing the presentation of a continuous range of stiffnesses in a single well without the confounding effect of differential cell migration. Using these nondurotactic gradient gels, stiffness-dependent hASC morphology, migration, and differentiation were studied. Finally, the mechanosensitive proteins YAP, Lamin A/C, Lamin B, MRTF-A, and MRTF-B were analyzed on these gradients, providing higher-resolution data on stiffness-dependent expression and localization.


Asunto(s)
Acrilamida/química , Resinas Acrílicas/química , Movimiento Celular/fisiología , Hidrogeles/química , Mecanotransducción Celular/fisiología , Células Madre/metabolismo , Adulto , Adhesión Celular/fisiología , Técnicas de Cultivo de Célula/métodos , Línea Celular , Módulo de Elasticidad/fisiología , Humanos , Polimerizacion
15.
Proc Natl Acad Sci U S A ; 113(12): 3185-90, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26944080

RESUMEN

Probing a wide range of cellular phenotypes in neurodevelopmental disorders using patient-derived neural progenitor cells (NPCs) can be facilitated by 3D assays, as 2D systems cannot entirely recapitulate the arrangement of cells in the brain. Here, we developed a previously unidentified 3D migration and differentiation assay in layered hydrogels to examine how these processes are affected in neurodevelopmental disorders, such as Rett syndrome. Our soft 3D system mimics the brain environment and accelerates maturation of neurons from human induced pluripotent stem cell (iPSC)-derived NPCs, yielding electrophysiologically active neurons within just 3 wk. Using this platform, we revealed a genotype-specific effect of methyl-CpG-binding protein-2 (MeCP2) dysfunction on iPSC-derived neuronal migration and maturation (reduced neurite outgrowth and fewer synapses) in 3D layered hydrogels. Thus, this 3D system expands the range of neural phenotypes that can be studied in vitro to include those influenced by physical and mechanical stimuli or requiring specific arrangements of multiple cell types.


Asunto(s)
Movimiento Celular , Hidrogeles , Células Madre Pluripotentes Inducidas/citología , Proteína 2 de Unión a Metil-CpG/fisiología , Neuronas/metabolismo , Humanos
16.
Circ Res ; 118(2): 296-310, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26838315

RESUMEN

Soluble morphogen gradients have long been studied in the context of heart specification and patterning. However, recent data have begun to challenge the notion that long-standing in vivo observations are driven solely by these gradients alone. Evidence from multiple biological models, from stem cells to ex vivo biophysical assays, now supports a role for mechanical forces in not only modulating cell behavior but also inducing it de novo in a process termed mechanotransduction. Structural proteins that connect the cell to its niche, for example, integrins and cadherins, and that couple to other growth factor receptors, either directly or indirectly, seem to mediate these changes, although specific mechanistic details are still being elucidated. In this review, we summarize how the wingless (Wnt), transforming growth factor-ß, and bone morphogenetic protein signaling pathways affect cardiomyogenesis and then highlight the interplay between each pathway and mechanical forces. In addition, we will outline the role of integrins and cadherins during cardiac development. For each, we will describe how the interplay could change multiple processes during cardiomyogenesis, including the specification of undifferentiated cells, the establishment of heart patterns to accomplish tube and chamber formation, or the maturation of myocytes in the fully formed heart.


Asunto(s)
Diferenciación Celular , Corazón/embriología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mecanotransducción Celular , Células Madre/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Cadherinas/metabolismo , Edad Gestacional , Humanos , Integrinas/metabolismo , Organogénesis , Estrés Mecánico , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt
17.
Circ Res ; 118(10): 1553-62, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27174949

RESUMEN

Unlike diet and exercise, which individuals can modulate according to their lifestyle, aging is unavoidable. With normal or healthy aging, the heart undergoes extensive vascular, cellular, and interstitial molecular changes that result in stiffer less compliant hearts that experience a general decline in organ function. Although these molecular changes deemed cardiac remodeling were once thought to be concomitant with advanced cardiovascular disease, they can be found in patients without manifestation of clinical disease. It is now mostly acknowledged that these age-related mechanical changes confer vulnerability of the heart to cardiovascular stresses associated with disease, such as hypertension and atherosclerosis. However, recent studies have aimed at differentiating the initial compensatory changes that occur within the heart with age to maintain contractile function from the maladaptive responses associated with disease. This work has identified new targets to improve cardiac function during aging. Spanning invertebrate to vertebrate models, we use this review to delineate some hallmarks of physiological versus pathological remodeling that occur in the cardiomyocyte and its microenvironment, focusing especially on the mechanical changes that occur within the sarcomere, intercalated disc, costamere, and extracellular matrix.


Asunto(s)
Enfermedades Cardiovasculares/genética , Drosophila/genética , Corazón/crecimiento & desarrollo , Miocitos Cardíacos/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Costameras/metabolismo , Modelos Animales de Enfermedad , Drosophila/metabolismo , Matriz Extracelular/metabolismo
18.
Biophys J ; 112(4): 736-745, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28256233

RESUMEN

Cancer cells within a tumor are heterogeneous and only a small fraction are able to form secondary tumors. Universal biological markers that clearly identify potentially metastatic cells are limited, which complicates isolation and further study. However, using physical rather than biological characteristics, we have identified Mg2+- and Ca2+-mediated differences in adhesion strength between metastatic and nonmetastatic mammary epithelial cell lines, which occur over concentration ranges similar to those found in tumor stroma. Metastatic cells exhibit remarkable heterogeneity in their adhesion strength under stromal-like conditions, unlike their nonmetastatic counterparts, which exhibit Mg2+- and Ca2+-insensitive adhesion. This heterogeneity is the result of increased sensitivity to Mg2+- and Ca2+-mediated focal adhesion disassembly in metastatic cells, rather than changes in integrin expression or focal adhesion phosphorylation. Strongly adherent metastatic cells exhibit less migratory behavior, similar to nonmetastatic cell lines but contrary to the unselected metastatic cell population. Adhesion strength heterogeneity was observed across multiple cancer cell lines as well as isogenically, suggesting that adhesion strength may serve as a general marker of metastatic cells.


Asunto(s)
Adhesión Celular , Metástasis de la Neoplasia , Línea Celular Tumoral , Movimiento Celular , Adhesiones Focales , Humanos
19.
J Cell Sci ; 128(10): 1961-8, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25908864

RESUMEN

Pluripotent embryonic stem cells (ESCs) exert low-traction forces on their niche in vitro whereas specification to definitive endoderm in vivo coincides with force-mediated motility, suggesting a differentiation-mediated switch. However, the onset of contractility and extent to which force-mediated integrin signaling regulates fate choices is not understood. To address the requirement of tractions forces for differentiation, we examined mouse embryonic stem cell (ESC) specification towards definitive endoderm on fibrillar fibronectin containing a deformation-sensitive FRET probe. Inhibiting contractility resulted in an increase in the observed fibronectin FRET intensity ratio but also decreased the amount of phosphorylated nuclear SMAD2, leading to reduced expression of the definitive endoderm marker SOX17. By contrast ESCs maintained in pluripotency medium did not exert significant tractions against the fibronectin matrix. When laminin-111 was added to fibrillar matrices to improve the efficiency of definitive endoderm induction, ESCs decreased their fibronectin traction forces in a laminin-dependent manner; blocking the laminin-binding α3-integrin restored fibronectin matrix deformation and reduced SOX17 expression and SMAD2 phosphorylation, probably because of compensation of inhibitory signaling from SMAD7 after 5 days in culture. These data imply that traction forces and integrin signaling are important regulators of early fate decisions in ESCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Endodermo/citología , Integrinas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/fisiología , Humanos , Ratones , Unión Proteica , Transducción de Señal
20.
Circ Res ; 114(2): e6-17, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24221941

RESUMEN

RATIONALE: Regulation of striated muscle contraction is achieved by Ca2+ -dependent steric modulation of myosin cross-bridge cycling on actin by the thin filament troponin-tropomyosin complex. Alterations in the complex can induce contractile dysregulation and disease. For example, mutations between or near residues 112 to 136 of cardiac troponin-T, the crucial TnT1 (N-terminal domain of troponin-T)-tropomyosin-binding region, cause cardiomyopathy. The Drosophila upheld(101) Glu/Lys amino acid substitution lies C-terminally adjacent to this phylogenetically conserved sequence. OBJECTIVE: Using a highly integrative approach, we sought to determine the molecular trigger of upheld(101) myofibrillar degeneration, to evaluate contractile performance in the mutant cardiomyocytes, and to examine the effects of the mutation on the entire Drosophila heart to elucidate regulatory roles for conserved TnT1 regions and provide possible mechanistic insight into cardiac dysfunction. METHODS AND RESULTS: Live video imaging of Drosophila cardiac tubes revealed that the troponin-T mutation prolongs systole and restricts diastolic dimensions of the heart, because of increased numbers of actively cycling myosin cross-bridges. Elevated resting myocardial stiffness, consistent with upheld(101) diastolic dysfunction, was confirmed by an atomic force microscopy-based nanoindentation approach. Direct visualization of mutant thin filaments via electron microscopy and 3-dimensional reconstruction resolved destabilized tropomyosin positioning and aberrantly exposed myosin-binding sites under low Ca2+ conditions. CONCLUSIONS: As a result of troponin-tropomyosin dysinhibition, upheld(101) hearts exhibited cardiac dysfunction and remodeling comparable to that observed during human restrictive cardiomyopathy. Thus, reversal of charged residues about the conserved tropomyosin-binding region of TnT1 may perturb critical intermolecular associations required for proper steric regulation, which likely elicits myopathy in our Drosophila model.


Asunto(s)
Cardiomiopatías/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Miofibrillas/metabolismo , Troponina T/metabolismo , Disfunción Ventricular/metabolismo , Función Ventricular , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Diástole , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestructura , Acoplamiento Excitación-Contracción , Femenino , Genotipo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Masculino , Microscopía Electrónica , Microscopía por Video , Datos de Secuencia Molecular , Mutación , Miofibrillas/efectos de los fármacos , Miofibrillas/ultraestructura , Fenotipo , Sístole , Tropomiosina/metabolismo , Troponina T/genética , Disfunción Ventricular/genética , Disfunción Ventricular/patología , Disfunción Ventricular/fisiopatología , Remodelación Ventricular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA