Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Biophys J ; 118(5): 1058-1066, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-31995740

RESUMEN

Detection of the transition between the two myosin isoforms α- and ß-myosin in living cardiomyocytes is essential for understanding cardiac physiology and pathology. In this study, the differences in symmetry of polarization spectra obtained from α- and ß-myosin in various mammalian ventricles and propylthiouracil-treated rats are explored through polarization-dependent second harmonic generation microscopy. Here, we report for the, to our knowledge, first time that α- and ß-myosin, as protein crystals, possess different symmetries: the former has C6 symmetry, and the latter has C3v. A single-sarcomere line scan further demonstrated that the differences in polarization-spectrum symmetry between α- and ß-myosin came from their head regions: the head and neck domains of α- and ß-myosin account for the differences in symmetry. In addition, the dynamic transition of the polarization spectrum from C6 to C3v line profile was observed in a cell culture in which norepinephrine induced an α- to ß-myosin transition.


Asunto(s)
Miosinas Cardíacas , Sarcómeros , Animales , Miocitos Cardíacos , Miosinas , Ratas , Miosinas Ventriculares
2.
Biomed Opt Express ; 10(7): 3183-3195, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31360597

RESUMEN

Detecting the structural changes caused by volume and pressure overload is critical to comprehending the mechanisms of physiologic and pathologic hypertrophy. This study explores the structural changes at the crystallographic level in myosin filaments in volume- and pressure-overloaded myocardia through polarization-dependent second harmonic generation microscopy. Here, for the first time, we report that the ratio of nonlinear susceptibility tensor components d33/d15 increased significantly in volume- and pressure-overloaded myocardial tissues compared with the ratio in normal mouse myocardial tissues. Through cell stretch experiments, we demonstrated that mechanical tension plays an important role in the increase of d33/d15 in volume- and pressure-overloaded myocardial tissues.

3.
Cytoskeleton (Hoboken) ; 73(5): 246-57, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27072949

RESUMEN

Myofibrils are the main protein structures that generate force in the beating heart. Myofibril disassembly is related to many physiological and pathological processes. This study investigated, in a cultured rat adult cardiomyocyte model, the effect of force imbalance on myofibril disassembly. The imbalance of forces that were exerted on Z-discs was induced by the synergistic effect of broken intercalated discs and actin-myosin interaction. Cardiomyocytes with well-preserved intercalated discs were isolated from adult rat ventricles. The ultrastructure of cardiomyocyte was observed using a customized two-photon excitation fluorescence and second harmonic generation imaging system. The contraction of cardiomyocytes was recorded with a high-speed CCD camera, and the movement of cellular components was analyzed using a contractile imaging assay technique. The cardiomyocyte dynamic remodeling process was recorded using a time-lapse imaging system. The role of actin-myosin interaction in myofibril disassembly was investigated by incubating cardiomyocytes with blebbistatin (25 µM). Results demonstrated that the hierarchical disassembly process of myofibrils was initiated from cardiomyocyte free ends where intercalated discs had broken, during which the desmin network near the free cell ends was destroyed to release single myofibrils. Analysis of force (based on a schematic model of cardiomyocytes connected at intercalated discs) suggests that breaking of intercalated discs caused force imbalance on both sides of the Z-discs adjacent to the cell ends due to actin-myosin interaction. The damaged intercalated discs and actin-myosin interaction induced force imbalance on both sides of the Z-discs, which played an important role in the hierarchical disassembly of myofibrils. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Actinas/metabolismo , Ventrículos Cardíacos/metabolismo , Modelos Biológicos , Miofibrillas/metabolismo , Miosinas/metabolismo , Animales , Ventrículos Cardíacos/citología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Ratas , Ratas Sprague-Dawley
4.
Biofabrication ; 8(1): 015012, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26844857

RESUMEN

Tunneling nanotubes (TNTs) are small membranous tubes of 50-1000 nm diameter observed to connect cells in culture. Transfer of subcellular organelles through TNTs was observed in vitro and in vivo, but the formation and significance of these structures is not well understood. A polydimethylsiloxane biochip-based coculture model was devised to constrain TNT orientation and explore both TNT-formation and TNT-mediated mitochondrial transfer. Two parallel microfluidic channels connected by an array of smaller microchannels enabled localization of stem cell and cardiomyocyte populations while allowing connections to form between them. Stem cells and cardiomyocytes were deposited in their respective microfluidic channels, and stem cell-cardiomyocyte pairs were formed via the microchannels. Formation of TNTs and transfer of stained mitochondria through TNTs was observed by 24 h real-time video recording. The data show that stem cells are 7.7 times more likely to initiate contact by initial extension of filopodia. By 24 h, 67% of nanotube connections through the microchannels are composed of cardiomyocyte membrane. Filopodial extension and retraction by stem cells draws an extension of TNTs from cardiomyocytes. MitoTracker staining shows that unidirectional transfer of mitochondria between stem cell-cardiomyocyte pairs invariably originates from stem cells. Control experiments with cardiac fibroblasts and cardiomyocytes show little nanotube formation between homotypic or mixed cell pairs and no mitochondrial transfer. These data identify a novel biological process, unidirectional mitochondrial transfer, mediated by heterotypic TNT connections. This suggests that the enhancement of cardiomyocyte function seen after stem-cell injection may be due to a bioenergetic stimulus provided by mitochondrial transfer.


Asunto(s)
Comunicación Celular/fisiología , Dispositivos Laboratorio en un Chip , Células Madre Mesenquimatosas/fisiología , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/fisiología , Animales , Técnicas de Cultivo de Célula/instrumentación , Extensiones de la Superficie Celular , Células Cultivadas , Diseño de Equipo , Análisis de Falla de Equipo , Células Madre Mesenquimatosas/ultraestructura , Mitocondrias Cardíacas/ultraestructura , Miocitos Cardíacos/ultraestructura , Nanotubos/ultraestructura , Ratas , Ratas Sprague-Dawley , Células Madre
5.
Sci Rep ; 6: 20674, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26861590

RESUMEN

An increase in mechanical load in the heart causes cardiac hypertrophy, either physiologically (heart development, exercise and pregnancy) or pathologically (high blood pressure and heart-valve regurgitation). Understanding cardiac hypertrophy is critical to comprehending the mechanisms of heart development and treatment of heart disease. However, the major molecular event that occurs during physiological or pathological hypertrophy is the dynamic process of sarcomeric addition, and it has not been observed. In this study, a custom-built second harmonic generation (SHG) confocal microscope was used to study dynamic sarcomeric addition in single neonatal CMs in a 3D culture system under acute, uniaxial, static, sustained stretch. Here we report, for the first time, live-cell observations of various modes of dynamic sarcomeric addition (and how these real-time images compare to static images from hypertrophic hearts reported in the literature): 1) Insertion in the mid-region or addition at the end of a myofibril; 2) Sequential addition with an existing myofibril as a template; and 3) Longitudinal splitting of an existing myofibril. The 3D cell culture system developed on a deformable substrate affixed to a stretcher and the SHG live-cell imaging technique are unique tools for real-time analysis of cultured models of hypertrophy.


Asunto(s)
Miocitos Cardíacos/citología , Miofibrillas/fisiología , Estrés Mecánico , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Análisis por Conglomerados , Microscopía Confocal , Mitocondrias/metabolismo , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Sarcómeros/fisiología
6.
Am J Physiol Heart Circ Physiol ; 308(9): H1112-25, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25595366

RESUMEN

Cardiac tissue slices are becoming increasingly popular as a model system for cardiac electrophysiology and pharmacology research and development. Here, we describe in detail the preparation, handling, and optical mapping of transmembrane potential and intracellular free calcium concentration transients (CaT) in ventricular tissue slices from guinea pigs and rabbits. Slices cut in the epicardium-tangential plane contained well-aligned in-slice myocardial cell strands ("fibers") in subepicardial and midmyocardial sections. Cut with a high-precision slow-advancing microtome at a thickness of 350 to 400 µm, tissue slices preserved essential action potential (AP) properties of the precutting Langendorff-perfused heart. We identified the need for a postcutting recovery period of 36 min (guinea pig) and 63 min (rabbit) to reach 97.5% of final steady-state values for AP duration (APD) (identified by exponential fitting). There was no significant difference between the postcutting recovery dynamics in slices obtained using 2,3-butanedione 2-monoxime or blebistatin as electromechanical uncouplers during the cutting process. A rapid increase in APD, seen after cutting, was caused by exposure to ice-cold solution during the slicing procedure, not by tissue injury, differences in uncouplers, or pH-buffers (bicarbonate; HEPES). To characterize intrinsic patterns of CaT, AP, and conduction, a combination of multipoint and field stimulation should be used to avoid misinterpretation based on source-sink effects. In summary, we describe in detail the preparation, mapping, and data analysis approaches for reproducible cardiac tissue slice-based investigations into AP and CaT dynamics.


Asunto(s)
Señalización del Calcio , Frío , Microtomía/métodos , Miocardio/metabolismo , Imagen de Colorante Sensible al Voltaje/métodos , Potenciales de Acción , Animales , Estimulación Cardíaca Artificial , Frío/efectos adversos , Femenino , Cobayas , Técnicas In Vitro , Cinética , Masculino , Perfusión , Conejos , Recuperación de la Función , Procesamiento de Señales Asistido por Computador , Supervivencia Tisular
7.
Exp Cell Res ; 330(1): 222-32, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25151177

RESUMEN

The cardiac basement membrane (BM), the highly organized layer of the extracellular matrix (ECM) on the external side of the sarcolemma, is mainly composed of laminin and collagen IV, which assemble a dense, well-organized network to surround the surface of each adult cardiomyocyte. The development of the cardiac BM plays a key role in organogenesis of the myocardium through interactions between sarcomeres and integrins. Because of the complicated structure of cardiac muscle fibers and lack of a proper investigation method, the detailed interactions among BM development, sarcomeric growth, and integrin expression remain unclear. In this study, freshly isolated 3-day neonatal cardiomyocytes (CMs) were cultured on aligned collagen, which mimics the in vivo ECM structure and induces neonatal CMs to grow into rod-like shapes. Then double fluorescence-immunostained laminin and α-actinin or integrin ß1 on neonatal CMs cultured 4-72 h were imaged using a confocal microscope, and the spatial relationship between laminin deposition and α-actinin expression was evaluated by colocalization analysis. At 4h, laminin was deposited around Z-bodies (dot-shaped α-actinin) and integrins; from 18-to-72 h, its gradual colocalization with Z-lines (line-shaped α-actinin) and integrins increased Pearson׳s coefficient; this indicates that development of the BM network from the neonatal stage to adulthood is closely related to sarcomeric formation via integrin-mediated interactions.


Asunto(s)
Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Sarcómeros/metabolismo , Actinina/genética , Actinina/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Corazón/crecimiento & desarrollo , Integrina beta1/genética , Integrina beta1/metabolismo , Laminina/genética , Laminina/metabolismo , Miocitos Cardíacos/citología , Ratas , Ratas Sprague-Dawley
8.
Biofabrication ; 6(4): 045003, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25215627

RESUMEN

The basement membrane (BM), a network of laminin and collagen IV, mechanically supports individual cells and directly mediates cell-cell and cell-extracellular matrix (ECM) interactions. For example, the BM network that tightly encloses each cardiomyocyte (CM) mediates the alignment of CMs with collagen I in the ECM. Additionally, the BM-laminin is involved in the formation of gap junctions (GJs), which regulate electrical coupling between two CMs in the myocardium. The role of BM in GJ maturation remains unclear because of the complicated in vivo structures and lack of an ideal in vitro culturing mode. In this study, our laser cell-micropatterning system was used to place two neonatal CMs (NCMs) in contact on an aligned collagen gel (ACG) to study the relationship between GJ maturation and BM development. The results of double immunofluorescence staining and confocal imaging showed that BM-laminin was deposited earlier than the formation of GJs in the intercellular space and that newly expressed connexin 43 clusters were preferentially assembled near the deposited BM structures. Eventually the BM network surrounded the GJs.


Asunto(s)
Membrana Basal/fisiología , Técnicas Citológicas/métodos , Uniones Comunicantes/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Animales , Ventrículos Cardíacos/citología , Rayos Láser , Microscopía Confocal , Microscopía Fluorescente , Ratas , Ratas Sprague-Dawley
9.
Ann Biomed Eng ; 42(6): 1148-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24577875

RESUMEN

In the heart muscle, each adult cardiomyocyte is enclosed by a basement membrane (BM). This innermost extracellular matrix is a layered assembly of laminin, collagen IV, glycoproteins, and proteoglycans. In this study, the role of the BM network in regulation of the electrical properties of neonatal cardiomyocytes (NCMs) cultured on an aligned collagen I gel was investigated using a multielectrode array (MEA). A laminin antibody was added to the culture medium for 48-120 h to conjugate newly secreted laminin. Then, morphology of the NCMs on an MEA was monitored using a phase contrast microscope, and the BM network that was immunocytostained for laminin was imaged using a fluorescence microscope. When the BM laminin was absent in this culture model, dramatic changes in NCM morphology were observed. Simultaneously, the MEA-recorded cardiac field potential showed changes compared to that from the control groups: The period of contraction shortened to 1/2 of that from the control groups, and the waveform of the calcium influx shifted from a flat plateau to a peak-like waveform, indicating that the electrical properties of the NCMs were closely related to the components and distribution of the BM network.


Asunto(s)
Membrana Basal/metabolismo , Fenómenos Electrofisiológicos , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Membrana Basal/citología , Células Cultivadas , Proteínas Musculares/metabolismo , Miocitos Cardíacos/citología , Ratas , Ratas Sprague-Dawley
10.
Biotechnol Lett ; 36(6): 1245-52, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24562408

RESUMEN

A technique to tailor-make pre-coated, pre-aligned bovine collagen fibrils, derived from neonatal cardiomyocytes, on the surface of a glass slide into a designated pattern is reported. The unwanted collagen-coated area was erased by a collagenase solution and the tailored area was retained by attaching a microfabricated polydimethylsiloxane stamp directly to the collagen-coated surface. Using this technique, collagen patterns with designated orientations and with clear pattern boundaries and defined shapes were fabricated.


Asunto(s)
Colágeno/metabolismo , Colagenasas/metabolismo , Microtecnología/métodos , Materiales Biocompatibles Revestidos , Humanos , Recién Nacido , Miocitos Cardíacos/química
11.
Proc SPIE Int Soc Opt Eng ; 89482014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25767692

RESUMEN

Second harmonic generation (SHG) microscopy is a new imaging technique used in sarcomeric-addition studies. However, during the early stage of cell culture in which sarcomeric additions occur, the neonatal cardiomyocytes that we have been working with are very sensitive to photodamage, the resulting high rate of cell death prevents systematic study of sarcomeric addition using a conventional SHG system. To address this challenge, we introduced use of the pulse-splitter system developed by Na Ji et al. in our two photon excitation fluorescence (TPEF) and SHG hybrid microscope. The system dramatically reduced photodamage to neonatal cardiomyocytes in early stages of culture, greatly increasing cell viability. Thus continuous imaging of live cardiomyocytes was achieved with a stronger laser and for a longer period than has been reported in the literature. The pulse splitter-based TPEF-SHG microscope constructed in this study was demonstrated to be an ideal imaging system for sarcomeric addition-related investigations of neonatal cardiomyocytes in early stages of culture.

12.
Methods Mol Biol ; 1075: 185-99, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24052352

RESUMEN

Detailed methods are provided for the preparation and confocal imaging of cardiac myocyte development and differentiation. Examples include protocols for the analysis of cultured myocytes as well as vibratome sections of hearts from embryonic and adult tissue. Techniques include routine labeling of F-actin with phalloidin as well as multiple labeling protocols for colocalization studies and cell volume analysis.


Asunto(s)
Diferenciación Celular/genética , Microscopía Confocal/métodos , Miocitos Cardíacos/ultraestructura , Actinas/ultraestructura , Adulto , Animales , Humanos , Ratones
13.
PLoS One ; 8(11): e79715, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223185

RESUMEN

Fibrillar collagen is the primary component of the cardiac interstitial extracellular matrix. This extracellular matrix undergoes dramatic changes from birth to adulthood and then into advanced age. As evidence, fibrillar collagen content was compared in sections from neonates, adult, and old hearts and was found to increase at each respective age. Cardiac fibroblasts are the principle cell type that produce and control fibrillar collagen content. To determine whether fibroblast production, processing, and deposition of collagen differed with age, primary cardiac fibroblasts from neonate, adult, and old mice were isolated and cultured in 3-dimensional (3D) fibrin gels. Fibroblasts from each age aligned in fibrin gels along points of tension and deposited extracellular matrix. By confocal microscopy, wild-type neonate fibroblasts appeared to deposit less collagen into fibrillar structures than fibroblasts from adults. However, by immunoblot analysis, differences in procollagen production and processing of collagen I were not detected in neonate versus adult fibroblasts. In contrast, fibroblasts from old mice demonstrated increased efficiency of procollagen processing coupled with decreased production of total collagen. SPARC is a collagen-binding protein previously shown to affect cardiac collagen deposition. Accordingly, in the absence of SPARC, less collagen appeared to be associated with fibroblasts of each age grown in fibrin gels. In addition, the increased efficiency of procollagen alpha 1(I) processing in old wild-type fibroblasts was not detected in old SPARC-null fibroblasts. Increased levels of fibronectin were detected in wild-type neonate fibroblasts over that of adult and old fibroblasts but not in SPARC-null neonate fibroblasts versus older ages. Immunostaining of SPARC overlapped with that of collagen I but not to that of fibronectin in 3D cultures. Hence, whereas increases in procollagen processing, influenced by SPARC expression, plausibly contribute to increased collagen deposition in old hearts, other cellular mechanisms likely affect differential collagen deposition by neonate fibroblasts.


Asunto(s)
Envejecimiento/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Miocardio/citología , Osteonectina/metabolismo , Animales , Técnicas de Cultivo de Célula , Colágeno Tipo I/análisis , Colágeno Tipo I/química , Fibroblastos/ultraestructura , Ratones , Procolágeno/química , Procolágeno/metabolismo , Solubilidad
14.
PLoS One ; 8(2): e56554, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23418583

RESUMEN

Understanding how stem cells interact with cardiomyocytes is crucial for cell-based therapies to restore the cardiomyocyte loss that occurs during myocardial infarction and other cardiac diseases. It has been thought that functional myocardial repair and regeneration could be regulated by stem cell-cardiomyocyte contact. However, because various contact modes (junction formation, cell fusion, partial cell fusion, and tunneling nanotube formation) occur randomly in a conventional coculture system, the particular regulation corresponding to a specific contact mode could not be analyzed. In this study, we used laser-patterned biochips to define cell-cell contact modes for systematic study of contact-mediated cellular interactions at the single-cell level. The results showed that the biochip design allows defined stem cell-cardiomyocyte contact-mode formation, which can be used to determine specific cellular interactions, including electrical coupling, mechanical coupling, and mitochondria transfer. The biochips will help us gain knowledge of contact-mediated interactions between stem cells and cardiomyocytes, which are fundamental for formulating a strategy to achieve stem cell-based cardiac tissue regeneration.


Asunto(s)
Comunicación Celular/fisiología , Rastreo Celular/métodos , Células Madre Mesenquimatosas/fisiología , Miocitos Cardíacos/fisiología , Animales , Animales Recién Nacidos , Fusión Celular , Membrana Celular/fisiología , Rastreo Celular/instrumentación , Células Cultivadas , Técnicas de Cocultivo , Colorantes Fluorescentes/química , Inmunohistoquímica , Indoles/química , Uniones Intercelulares/fisiología , Rayos Láser , Células Madre Mesenquimatosas/citología , Microscopía Confocal , Microscopía Fluorescente , Mitocondrias/fisiología , Miocitos Cardíacos/citología , Ratas , Reproducibilidad de los Resultados , Análisis de la Célula Individual/instrumentación , Análisis de la Célula Individual/métodos
15.
Cardiovasc Res ; 97(2): 262-70, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23118131

RESUMEN

AIMS: Understanding myofibrillogenesis is essential for elucidating heart muscle formation, development, and remodelling in response to physiological stimulation. Here, we report the dynamic assembly process of contractile myosin filaments onto myofibrils in a live cardiomyocyte culture during myofibrillogenesis. METHODS AND RESULTS: Utilizing a custom-built, two-photon excitation fluorescence and second harmonic generation imaging system equipped with an on-stage incubator, we observed new sarcomere additions in rat neonatal cardiomyocytes during 10 h of on-stage incubation. The new sarcomere additions occurred at the side of existing myofibrils, where we observed mature myofibrils acting as templates, or at the interstice of several separated myofibrils. CONCLUSIONS: During sarcomeric addition, myosin filaments are assembled onto the premyofibril laterally. This lateral addition, which proceeds stepwise along the axial direction, plays an important role in the accumulation of Z-bodies to form mature Z-disks and in the regulation of sarcomeric alignment during maturation.


Asunto(s)
Desarrollo de Músculos , Miocitos Cardíacos/ultraestructura , Miosinas/química , Animales , Animales Recién Nacidos , Microscopía Confocal , Miofibrillas/ultraestructura , Ratas , Ratas Sprague-Dawley , Sarcómeros/ultraestructura
16.
Artículo en Inglés | MEDLINE | ID: mdl-24527266

RESUMEN

Mesenchymal stem cells (MSCs) have been cited as contributors to heart repair through cardiogenic differentiation and multiple cellular interactions, including the paracrine effect, cell fusion, and mechanical and electrical couplings. Due to heart-muscle complexity, progress in the development of knowledge concerning the role of MSCs in cardiac repair is heavily based on MSC-cardiomyocyte coculture. In conventional coculture systems, however, the in vivo cardiac muscle structure, in which rod-shaped cells are connected end-to-end, is not sustained; instead, irregularly shaped cells spread randomly, resulting in randomly distributed cell junctions. Consequently, contact-mediated cell-cell interactions (e.g., the electrical triggering signal and the mechanical contraction wave that propagate through MSC-cardiomyocyte junctions) occur randomly. Thus, the data generated on the beneficial effects of MSCs may be irrelevant to in vivo biological processes. In this study, we explored whether cardiomyocyte alignment, the most important phenotype, is relevant to stem cell cardiogenic differentiation. Here, we report (i) the construction of a laser-patterned, biochip-based, stem cell-cardiomyocyte coculture model with controlled cell alignment; and (ii) single-cell-level data on stem cell cardiogenic differentiation under in vivo-like cardiomyocyte alignment conditions.

17.
Cell Mol Bioeng ; 5(3): 327-336, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23139730

RESUMEN

Normal cardiomyocytes are highly dependent on the functional expression of ion channels to form action potentials and electrical coupling with other cells. To fully determine the scientific and therapeutic potential of stem cells for cardiovascular-disease treatment, it is necessary to assess comprehensively the regulation of stem-cell electrical properties during stem cell-cardiomyocyte interaction. It has been reported in the literature that contact with native cardiomyocytes induced and regulated stem-cell cardiogenic differentiation. However, in conventional cell-culture models, the importance of cell-cell contact for stem-cell functional coupling with cardiomyocytes has not been elucidated due to insufficient control of the cell-contact mode of individual cells. Using microfabrication and laser-guided cell micropatterning techniques, we created two biochips with contact-promotive and -preventive microenvironments to systematically study the effect of contact on cardiogenic regulation of stem-cell electrical properties. In contact-promotive biochips, connexin 43 expression was upregulated and relocated to the junction area between one stem cell and one cardiomyocyte. Only stem cells in contact with cardiomyocytes were induced by adjacent cardiomyocytes to acquire electrophysiological properties for action-potential formation similar to that of a cardiomyocyte.

18.
Am J Pathol ; 181(4): 1226-35, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22954422

RESUMEN

Cardiac hypertrophy, whether pathological or physiological, induces a variety of additional morphological and physiological changes in the heart, including altered contractility and hemodynamics. Events exacerbating these changes are documented during later stages of hypertrophy (usually termed pathological hypertrophy). Few studies document the morphological and physiological changes during early physiological hypertrophy. We define acute cardiac remodeling events in response to transverse aortic constriction (TAC), including temporal changes in hypertrophy, collagen deposition, capillary density, and the cell populations responsible for these changes. Cardiac hypertrophy induced by TAC in mice was detected 2 days after surgery (as measured by heart weight, myocyte width, and wall thickness) and peaked by day 7. Picrosirius staining revealed increased collagen deposition 7 days after TAC; immunostaining and flow cytometry indicated a concurrent increase in fibroblasts. The findings correlated with angiogenesis in TAC hearts; a decrease in capillary density was observed at day 2, with recovery to sham-surgery levels by day 7. Increased pericyte levels, which were observed 2 days after TAC, may mediate this angiogenic transition. Gene expression suggests a coordinated response in growth, extracellular matrix, and angiogenic factors to mediate the observed morphological changes. Our data demonstrate that morphological changes in response to cardiovascular injury occur rapidly, and the present findings allow correlation of specific events that facilitate these changes.


Asunto(s)
Cardiomegalia/patología , Miocardio/patología , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aorta Torácica/fisiopatología , Aorta Torácica/cirugía , Cardiomegalia/fisiopatología , Proliferación Celular , Colágeno/metabolismo , Constricción Patológica , Modelos Animales de Enfermedad , Matriz Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/ultraestructura , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Coloración y Etiquetado , Remodelación Ventricular
20.
Microsc Microanal ; 18(1): 107-14, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22152112

RESUMEN

Normal cardiac function is maintained through dynamic interactions of cardiac cells with each other and with the extracellular matrix. These interactions are important for remodeling during cardiac growth and pathophysiological conditions. However, the precise mechanisms of these interactions remain unclear. In this study we examined the importance of desmoplakin (DSP) in cardiac cell-cell interactions. Cell-cell communication in the heart requires the formation and preservation of cell contacts by cell adhesion junctions called desmosome-like structures. A major protein component of this complex is DSP, which plays a role in linking the cytoskeletal network to the plasma membrane. Our laboratory previously generated a polyclonal antibody (1611) against the detergent soluble fraction of cardiac fibroblast plasma membrane. In attempting to define which proteins 1611 recognizes, we performed two-dimensional electrophoresis and identified DSP as one of the major proteins recognized by 1611. Immunoprecipitation studies demonstrated that 1611 was able to directly pulldown DSP. We also demonstrate that 1611 and anti-DSP antibodies co-localize in whole heart sections. Finally, using a three-dimensional in vitro cell-cell interaction assay, we demonstrate that 1611 can inhibit cell-cell interactions. These data indicate that DSP is an important protein for cell-cell interactions and affects a variety of cellular functions, including cytokine secretion.


Asunto(s)
Comunicación Celular , Desmoplaquinas/metabolismo , Fibroblastos/fisiología , Corazón/fisiología , Células Musculares/fisiología , Animales , Células Cultivadas , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA