Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 560: 139-145, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-33989905

RESUMEN

Drug screening and disease modelling for skeletal muscle related pathologies would strongly benefit from the integration of myogenic cells derived from human pluripotent stem cells within miniaturized cell culture devices, such as microfluidic platform. Here, we identified the optimal culture conditions that allow direct differentiation of human pluripotent stem cells in myogenic cells within microfluidic devices. Myogenic cells are efficiently derived from both human embryonic (hESC) or induced pluripotent stem cells (hiPSC) in eleven days by combining small molecules and non-integrating modified mRNA (mmRNA) encoding for the master myogenic transcription factor MYOD. Our work opens new perspective for the development of patient-specific platforms in which a one-step myogenic differentiation could be used to generate skeletal muscle on-a-chip.


Asunto(s)
Diferenciación Celular/genética , Fibras Musculares Esqueléticas/citología , Proteína MioD/genética , Células Madre Pluripotentes/citología , Línea Celular , Humanos , Dispositivos Laboratorio en un Chip , Mesodermo/citología , Desarrollo de Músculos , ARN Mensajero , Transfección
2.
PLoS One ; 15(5): e0232081, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32374763

RESUMEN

The reproduction of reliable in vitro models of human skeletal muscle is made harder by the intrinsic 3D structural complexity of this tissue. Here we coupled engineered hydrogel with 3D structural cues and specific mechanical properties to derive human 3D muscle constructs ("myobundles") at the scale of single fibers, by using primary myoblasts or myoblasts derived from embryonic stem cells. To this aim, cell culture was performed in confined, laminin-coated micrometric channels obtained inside a 3D hydrogel characterized by the optimal stiffness for skeletal muscle myogenesis. Primary myoblasts cultured in our 3D culture system were able to undergo myotube differentiation and maturation, as demonstrated by the proper expression and localization of key components of the sarcomere and sarcolemma. Such approach allowed the generation of human myobundles of ~10 mm in length and ~120 µm in diameter, showing spontaneous contraction 7 days after cell seeding. Transcriptome analyses showed higher similarity between 3D myobundles and skeletal signature, compared to that found between 2D myotubes and skeletal muscle, mainly resulting from expression in 3D myobundles of categories of genes involved in skeletal muscle maturation, including extracellular matrix organization. Moreover, imaging analyses confirmed that structured 3D culture system was conducive to differentiation/maturation also when using myoblasts derived from embryonic stem cells. In conclusion, our structured 3D model is a promising tool for modelling human skeletal muscle in healthy and diseases conditions.


Asunto(s)
Técnicas de Cultivo de Célula , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Cultivadas , Dimetilpolisiloxanos/química , Humanos , Hidrogeles/química , Ensayo de Materiales , Ratones , Modelos Biológicos , Conformación Molecular , Desarrollo de Músculos , Músculo Esquelético/fisiología , Mioblastos/citología , Mioblastos/fisiología , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos , Ingeniería de Tejidos/instrumentación , Ingeniería de Tejidos/métodos
3.
Mater Sci Eng C Mater Biol Appl ; 96: 625-634, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30606574

RESUMEN

Hydrogels are an increasingly attractive choice in the fields of regenerative medicine, wound care and tissue engineering as important forms of bio-scaffolds. For many clinical needs, injectable in situ crosslinkable hydrogels are strongly preferred, due to treatment effectiveness and ease of use. In this study, hyaluronic acid (HA), containing side-arms linked to photo-active coumarin moieties, was used for the preparation of wall-to-wall hydrogels. This photocrosslinkable HA, hereafter called HA-TEG-coumarin, produces colourless aqueous solutions that solidify upon near-UV irradiation (at a specific wavelength of 365 nm) via a clean [2 + 2] photocycloaddition reaction, without by-products formation. The crosslinking event, a robust and non-cytotoxic process, does not require catalysts or radical initiators: in the field of hyaluronan photocrosslinking, this innovative feature is significant to ensure the whole biocompatibility and to avoid collateral reactions. Mechanical and rheological tests showed that hyaluronan derivatives became hydrogels after 3-5 min of irradiation, with average values for bulk and surface elastic moduli of about 32 kPa and 193 kPa, respectively. Fluorescence recovery after photobleaching (FRAP) assay showed that the hydrogels are porous and allow a good permeation for nutrients and growth factors. Cell metabolism and proliferation assays revealed that hydrogel-encapsulated fibroblasts maintained their viability and that HA-TEG-coumarin sustained the proliferation of non-adherent myoblasts. For all of these reasons and thanks to a safe free-radical approach, this novel hyaluronan coumarin derivative could be a good candidate for tissue engineering and regenerative medicine applications.


Asunto(s)
Cumarinas/química , Reactivos de Enlaces Cruzados/química , Ácido Hialurónico/química , Hidrogeles/química , Procesos Fotoquímicos , Ingeniería de Tejidos , Rayos Ultravioleta , Animales , Línea Celular , Humanos , Ensayo de Materiales , Ratones
4.
Ann Biomed Eng ; 47(3): 852-865, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30569242

RESUMEN

Cell junctions play an important role in coordinating intercellular communication and intracellular ultrastructures, with desmosomes representing the mechanical component of such intercellular connections. Mutations to desmosomal component proteins compromise both inter- and intracellular signalling and correlate with severe diseases like arrhythmogenic cardiomyopathy (AC), with pathological phenotypes in tissues subjected to intense mechanical stimuli (skin and heart). Here, we explore the consequences of dysfunctional desmosomes in one line of induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) derived from an AC patient with a homozygous pathogenic mutation in desmosomal component protein plakophilin-2 (PKP2). We specifically aim at investigating the response to mechanical stress in an AC-pathological setting. To this aim, we aligned hiPS-CMs on stretchable patterned substrates to mimic the cardiac functional syncytium and compared transcriptomic profiles of PKP2-mutated hiPS-CMs and healthy controls. AC-CMs display altered transcription towards a pro-fibrotic gene expression program, and concurrent dysregulation of gene sets closely associated with cell-to-cell connections. By integrating the culture substrate with a macroscopic stretching setup able to accurately apply cyclic uniaxial elongation, we show how response to mechanical loads in AC-CMs deviates from the canonical mechanical-stress response observed in healthy-CMs.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Cardiomiopatías/fisiopatología , Miocitos Cardíacos/fisiología , Tipificación del Cuerpo , Desmosomas/fisiología , Análisis de Elementos Finitos , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Estimulación Física , Placofilinas/genética , Estrés Mecánico
5.
Stem Cell Res ; 25: 107-114, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29125993

RESUMEN

Physical cues are major determinants of cellular phenotype and evoke physiological and pathological responses on cell structure and function. Cellular models aim to recapitulate basic functional features of their in vivo counterparts or tissues in order to be of use in in vitro disease modeling or drug screening and testing. Understanding how culture systems affect in vitro development of human pluripotent stem cell (hPSC)-derivatives allows optimization of cellular human models and gives insight in the processes involved in their structural organization and function. In this work, we show involvement of the mechanotransduction pathway RhoA/ROCK in the structural reorganization of hPSC-derived cardiomyocytes after adhesion plating. These structural changes have a major impact on the intracellular localization of SERCA2 pumps and concurrent improvement in calcium cycling. The process is triggered by cell interaction with the culture substrate, which mechanical cues drive sarcomeric alignment and SERCA2a spreading and relocalization from a perinuclear to a whole-cell distribution. This structural reorganization is mediated by the mechanical properties of the substrate, as shown by the process failure in hPSC-CMs cultured on soft 4kPa hydrogels as opposed to physiologically stiff 16kPa hydrogels and glass. Finally, pharmacological inhibition of Rho-associated protein kinase (ROCK) by different compounds identifies this specific signaling pathway as a major player in SERCA2 localization and the associated improvement in hPSC-CMs calcium handling ability in vitro.


Asunto(s)
Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Humanos , Mecanotransducción Celular/genética , Mecanotransducción Celular/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
6.
World J Radiol ; 9(1): 10-16, 2017 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-28144402

RESUMEN

AIM: To investigate the role of contrast enhanced ultrasound (CEUS) in evaluating patients with renal function impairment (RFI) showing: (1) acute renal failure (ARF) of suspicious vascular origin; or (2) suspicious renal lesions. METHODS: We retrospectively evaluated patients addressed to CEUS over an eight years period to rule-out vascular causes of ARF (first group of 50 subjects) or assess previously found suspicious renal lesions (second group of 41 subjects with acute or chronic RFI). After preliminary grey-scale and color Doppler investigation, each kidney was investigated individually with CEUS, using 1.2-2.4 mL of a sulfur hexafluoride-filled microbubble contrast agent. Image analysis was performed in consensus by two readers who reviewed digital clips of CEUS. We calculated the detection rate of vascular abnormalities in the first group and performed descriptive statistics of imaging findings for the second group. RESULTS: In the first group, CEUS detected renal infarction or cortical ischemia in 18/50 patients (36%; 95%CI: 23.3-50.9) and 1/50 patients (2%; 95%CI: 0.1-12), respectively. The detection rate of infarction was significantly higher (P = 0.0002; McNemar test) compared to color Doppler ultrasonography (10%). No vascular causes of ARF were identified in the remaining 31/50 patients (62%). In the second group, CEUS detected 41 lesions on 39 patients, allowing differentiation between solid lesions (21/41; 51.2%) vs complex cysts (20/41; 48.8%), and properly addressing 15/39 patients to intervention when feasible based on clinical conditions (surgery and cryoablation in 13 and 2 cases, respectively). Cysts were categorized Bosniak II, IIF, III and IV in 8, 5, 4 and 3 cases, respectively. In the remaining two patients, CEUS found 1 pseudolesion and 1 subcapsular hematoma. CONCLUSION: CEUS showed high detection rate of renal perfusion abnormalities in patients with ARF, influencing the management of patients with acute or chronic RFI and renal masses throughout their proper characterization.

7.
Langmuir ; 32(46): 12190-12201, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27643958

RESUMEN

The mechanical activity of cardiomyocytes is the result of a process called excitation-contraction coupling (ECC). A membrane depolarization wave induces a transient cytosolic calcium concentration increase that triggers activation of calcium-sensitive contractile proteins, leading to cell contraction and force generation. An experimental setup capable of acquiring simultaneously all ECC features would have an enormous impact on cardiac drug development and disease study. In this work, we develop a microengineered elastomeric substrate with tailor-made surface chemistry to measure simultaneously the uniaxial contraction force and the calcium transients generated by single human cardiomyocytes in vitro. Microreplication followed by photocuring is used to generate an array consisting of elastomeric micropillars. A second photochemical process is employed to spatially control the surface chemistry of the elastomeric pillar. As result, human embryonic stem cell-derived cardiomyocytes (hESC-CMs) can be confined in rectangular cell-adhesive areas, which induce cell elongation and promote suspended cell anchoring between two adjacent micropillars. In this end-to-end conformation, confocal fluorescence microscopy allows simultaneous detection of calcium transients and micropillar deflection induced by a single-cell uniaxial contraction force. Computational finite elements modeling (FEM) and 3D reconstruction of the cell-pillar interface allow force quantification. The platform is used to follow calcium dynamics and contraction force evolution in hESC-CMs cultures over the course of several weeks. Our results show how a biomaterial-based platform can be a versatile tool for in vitro assaying of cardiac functional properties of single-cell human cardiomyocytes, with applications in both in vitro developmental studies and drug screening on cardiac cultures.


Asunto(s)
Calcio/química , Elastómeros , Células Madre Embrionarias Humanas/citología , Miocitos Cardíacos/citología , Diferenciación Celular , Células Cultivadas , Humanos , Fenómenos Mecánicos , Microtecnología
8.
Stem Cells Transl Med ; 5(12): 1676-1683, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27502519

RESUMEN

: Restoration of the protein dystrophin on muscle membrane is the goal of many research lines aimed at curing Duchenne muscular dystrophy (DMD). Results of ongoing preclinical and clinical trials suggest that partial restoration of dystrophin might be sufficient to significantly reduce muscle damage. Different myogenic progenitors are candidates for cell therapy of muscular dystrophies, but only satellite cells and pericytes have already entered clinical experimentation. This study aimed to provide in vitro quantitative evidence of the ability of mesoangioblasts to restore dystrophin, in terms of protein accumulation and distribution, within myotubes derived from DMD patients, using a microengineered model. We designed an ad hoc experimental strategy to miniaturize on a chip the standard process of muscle regeneration independent of variables such as inflammation and fibrosis. It is based on the coculture, at different ratios, of human dystrophin-positive myogenic progenitors and dystrophin-negative myoblasts in a substrate with muscle-like physiological stiffness and cell micropatterns. Results showed that both healthy myoblasts and mesoangioblasts restored dystrophin expression in DMD myotubes. However, mesoangioblasts showed unexpected efficiency with respect to myoblasts in dystrophin production in terms of the amount of protein produced (40% vs. 15%) and length of the dystrophin membrane domain (210-240 µm vs. 40-70 µm). These results show that our microscaled in vitro model of human DMD skeletal muscle validated previous in vivo preclinical work and may be used to predict efficacy of new methods aimed at enhancing dystrophin accumulation and distribution before they are tested in vivo, reducing time, costs, and variability of clinical experimentation. SIGNIFICANCE: This study aimed to provide in vitro quantitative evidence of the ability of human mesoangioblasts to restore dystrophin, in terms of protein accumulation and distribution, within myotubes derived from patients with Duchenne muscular dystrophy (DMD), using a microengineered model. An ad hoc experimental strategy was designed to miniaturize on a chip the standard process of muscle regeneration independent of variables such as inflammation and fibrosis. This microscaled in vitro model, which validated previous in vivo preclinical work, revealed that mesoangioblasts showed unexpected efficiency as compared with myoblasts in dystrophin production. Consequently, this model may be used to predict efficacy of new drugs or therapies aimed at enhancing dystrophin accumulation and distribution before they are tested in vivo.


Asunto(s)
Diferenciación Celular , Distrofina/metabolismo , Modelos Biológicos , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Mioblastos/metabolismo , Mioblastos/patología , Donantes de Tejidos , Bioensayo , Técnicas de Cocultivo , Distrofina/química , Humanos , Análisis por Micromatrices , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/patología , Dominios Proteicos , Reproducibilidad de los Resultados
9.
Nat Methods ; 13(5): 446-52, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27088312

RESUMEN

We report that the efficiency of reprogramming human somatic cells to induced pluripotent stem cells (hiPSCs) can be dramatically improved in a microfluidic environment. Microliter-volume confinement resulted in a 50-fold increase in efficiency over traditional reprogramming by delivery of synthetic mRNAs encoding transcription factors. In these small volumes, extracellular components of the TGF-ß and other signaling pathways exhibited temporal regulation that appears critical to acquisition of pluripotency. The high quality and purity of the resulting hiPSCs (µ-hiPSCs) allowed direct differentiation into functional hepatocyte- and cardiomyocyte-like cells in the same platform without additional expansion.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular/genética , Células Madre Pluripotentes Inducidas/citología , Microfluídica/métodos , Células Cultivadas , Fibroblastos/citología , Humanos , ARN Mensajero/genética
10.
Curr Opin Biotechnol ; 25: 45-50, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24484880

RESUMEN

Organ-on-chip systems integrate microfluidic technology and living cells to study human physiology and pathophysiology. These human in vitro models are promising substitutes for animal testing, and their small scale enables precise control of culture conditions and high-throughput experiments, which would not be economically sustainable on a macroscopic level. Multiple sources of biological material are used in the development of organ-on-chips, from biopsies to stem cells. Each source has its own peculiarities and technical requirements for integration into microfluidic chips, and is suitable for specific applications. While a biopsy is the tissue of choice for the biomimetic response to ageing, induced pluripotent stem cells hold great promise for the study of genetic-related disease pathogenesis, and primary cultures can fill the gap.


Asunto(s)
Microfluídica/métodos , Animales , Técnicas de Cultivo de Célula , Humanos , Modelos Biológicos , Técnicas de Cultivo de Órganos , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA