Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 2281, 2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33500551

RESUMEN

In vitro neuronal models are essential for studying neurological physiology, disease mechanisms and potential treatments. Most in vitro models lack controlled vasculature, despite its necessity in brain physiology and disease. Organ-on-chip models offer microfluidic culture systems with dedicated micro-compartments for neurons and vascular cells. Such multi-cell type organs-on-chips can emulate neurovascular unit (NVU) physiology, however there is a lack of systematic data on how individual cell types are affected by culturing on microfluidic systems versus conventional culture plates. This information can provide perspective on initial findings of studies using organs-on-chip models, and further optimizes these models in terms of cellular maturity and neurovascular physiology. Here, we analysed the transcriptomic profiles of co-cultures of human induced pluripotent stem cell (hiPSC)-derived neurons and rat astrocytes, as well as one-day monocultures of human endothelial cells, cultured on microfluidic chips. For each cell type, large gene expression changes were observed when cultured on microfluidic chips compared to conventional culture plates. Endothelial cells showed decreased cell division, neurons and astrocytes exhibited increased cell adhesion, and neurons showed increased maturity when cultured on a microfluidic chip. Our results demonstrate that culturing NVU cell types on microfluidic chips changes their gene expression profiles, presumably due to distinct surface-to-volume ratios and substrate materials. These findings inform further NVU organ-on-chip model optimization and support their future application in disease studies and drug testing.


Asunto(s)
Astrocitos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Madre Pluripotentes Inducidas/citología , Microfluídica , Neuronas/metabolismo , Transcriptoma/genética , Animales , Adhesión Celular/genética , Diferenciación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Neuronas/citología , Ratas
2.
Microsyst Nanoeng ; 6: 107, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-34567716

RESUMEN

Microfluidic systems enable automated and highly parallelized cell culture with low volumes and defined liquid dosing. To achieve this, systems typically integrate all functions into a single, monolithic device as a "one size fits all" solution. However, this approach limits the end users' (re)design flexibility and complicates the addition of new functions to the system. To address this challenge, we propose and demonstrate a modular and standardized plug-and-play fluidic circuit board (FCB) for operating microfluidic building blocks (MFBBs), whereby both the FCB and the MFBBs contain integrated valves. A single FCB can parallelize up to three MFBBs of the same design or operate MFBBs with entirely different architectures. The operation of the MFBBs through the FCB is fully automated and does not incur the cost of an extra external footprint. We use this modular platform to control three microfluidic large-scale integration (mLSI) MFBBs, each of which features 64 microchambers suitable for cell culturing with high spatiotemporal control. We show as a proof of principle that we can culture human umbilical vein endothelial cells (HUVECs) for multiple days in the chambers of this MFBB. Moreover, we also use the same FCB to control an MFBB for liquid dosing with a high dynamic range. Our results demonstrate that MFBBs with different designs can be controlled and combined on a single FCB. Our novel modular approach to operating an automated microfluidic system for parallelized cell culture will enable greater experimental flexibility and facilitate the cooperation of different chips from different labs.

3.
Sci Rep ; 8(1): 13524, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30202042

RESUMEN

We present a novel and highly reproducible process to fabricate transferable porous PDMS membranes for PDMS-based Organs-on-Chips (OOCs) using microelectromechanical systems (MEMS) fabrication technologies. Porous PDMS membranes with pore sizes down to 2.0 µm in diameter and a wide porosity range (2-65%) can be fabricated. To overcome issues normally faced when using replica moulding and extend the applicability to most OOCs and improve their scalability and reproducibility, the process includes a sacrificial layer to easily transfer the membranes from a silicon carrier to any PDMS-based OOC. The highly reliable fabrication and transfer method does not need of manual handling to define the pore features (size, distribution), allowing very thin (<10 µm) functional membranes to be transferred at chip level with a high success rate (85%). The viability of cell culturing on the porous membranes was assessed by culturing two different cell types on transferred membranes in two different OOCs. Human umbilical endothelial cells (HUVEC) and MDA-MB-231 (MDA) cells were successfully cultured confirming the viability of cell culturing and the biocompatibility of the membranes. The results demonstrate the potential of controlling the porous membrane features to study cell mechanisms such as transmigrations, monolayer formation, and barrier function. The high control over the membrane characteristics might consequently allow to intentionally trigger or prevent certain cellular responses or mechanisms when studying human physiology and pathology using OOCs.


Asunto(s)
Órganos Artificiales , Dimetilpolisiloxanos/química , Dispositivos Laboratorio en un Chip , Membranas Artificiales , Microfluídica/métodos , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Supervivencia Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ensayo de Materiales , Porosidad , Reproducibilidad de los Resultados
4.
Biochem Biophys Res Commun ; 497(4): 1135-1141, 2018 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-28153730

RESUMEN

Multi electrode arrays (MEAs) are increasingly used to detect external field potentials in electrically active cells. Recently, in combination with cardiomyocytes derived from human (induced) pluripotent stem cells they have started to become a preferred tool to examine newly developed drugs for potential cardiac toxicity in pre-clinical safety pharmacology. The most important risk parameter is proarrhythmic activity in cardiomyocytes which can cause sudden cardiac death. Whilst MEAs can provide medium- to high- throughput noninvasive assay platform, the translation of a field potential to cardiac action potential (normally measured by low-throughput patch clamp) is complex so that accurate assessment of drug risk to the heart is in practice still challenging. To address this, we used computational simulation to study the theoretical relationship between aspects of the field potential and the underlying cardiac action potential. We then validated the model in both primary mouse- and human pluripotent (embryonic) stem cell-derived cardiomyocytes showing that field potentials measured in MEAs could be converted to action potentials that were essentially identical to those determined directly by electrophysiological patch clamp. The method significantly increased the amount of information that could be extracted from MEA measurements and thus combined the advantages of medium/high throughput with more informative readouts. We believe that this will benefit the analysis of drug toxicity screening of cardiomyocytes using in time and accuracy.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Miocitos Cardíacos/efectos de los fármacos , Potenciales de Acción , Animales , Simulación por Computador , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Microelectrodos , Modelos Teóricos , Células Madre Pluripotentes/citología
5.
Neth Heart J ; 21(7-8): 367-71, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23864481

RESUMEN

Sarcomeres are the smallest contractile units of heart and skeletal muscles and are essential for generation and propagation of mechanical force in these striated muscles. During the last decades it has become increasingly clear that components of sarcomeres also play a fundamental role in signal transduction in physiological and pathophysiological conditions. Mutations or misexpression of both sarcomeric contractile and non-contractile proteins have been associated with a variety of cardiac diseases. Moreover, re-expression of foetal sarcomeric proteins or isoforms during cardiac disease can be observed, emphasising the importance of understanding signalling in sarcomeres in both development and disease. The prospective of pharmacological intervention at the level of the sarcomere is now emerging and may lead to novel therapeutic strategies for the treatment of cardiac and skeletal muscle diseases. These aspects will be discussed in this brief review and recent findings, which led to novel insights into the role of the sarcomeric cytoskeleton in muscle development and disease, will be highlighted.

6.
Stem Cell Res ; 10(1): 48-56, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23089628

RESUMEN

Unexpected induction of arrhythmias in the heart is still one of the major risks of new drugs despite recent improvements in cardiac safety assays. Here we address this in a novel emerging assay system. Eleven reference compounds were administrated to spontaneously beating clusters of cardiomyocytes from human pluripotent stem cells (hPSC-CM) and the responses determined using multi-electrode arrays. Nine showed clear dose-dependence effects on field potential (FP) duration. Of these, the Ca(2+) channel blockers caused profound shortening of action potentials, whereas the classical hERG blockers, like dofetilide and d,l-sotalol, induced prolongation, as expected. Unexpectedly, two potent blockers of the slow component of the delayed rectifier potassium current (I(Ks)), HMR1556 and JNJ303, had only minor effects on the extracellular FP of wild-type hPSC-CM despite evidence of functional I(Ks) channels. These compounds were therefore re-evaluated under conditions that mimicked reduced "repolarization reserve," a parameter reflecting the capacity of cardiomyocytes to repolarize and a strong risk factor for the development of ventricular arrhythmias. Strikingly, in both pharmacological and genetic models of diminished repolarization reserve, HMR1556 and JNJ03 strongly increased the FP duration. These profound effects indicate that I(Ks) plays an important role in limiting action potential prolongation when repolarization reserve is attenuated. The findings have important clinical implications and indicate that enhanced sensitization to repolarization-prolonging compounds through pharmacotherapy or genetic predisposition should be taken into account when assessing drug safety.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Miocitos Cardíacos/citología , Células Madre Pluripotentes/citología , Bloqueadores de los Canales de Potasio/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Miocitos Cardíacos/efectos de los fármacos , Técnicas de Placa-Clamp , Fenetilaminas/farmacología , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Sotalol/farmacología , Sulfonamidas/farmacología
7.
Cell Mol Life Sci ; 67(2): 277-90, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19844658

RESUMEN

Transplantation of human embryonic stem cell-derived cardiomyocytes (hESC-CM) for cardiac regeneration is hampered by the formation of fibrotic tissue around the grafts, preventing electrophysiological coupling. Investigating this process, we found that: (1) beating hESC-CM in vitro are embedded in collagens, laminin and fibronectin, which they bind via appropriate integrins; (2) after transplantation into the mouse heart, hESC-CM continue to secrete collagen IV, XVIII and fibronectin; (3) integrin expression on hESC-CM largely matches the matrix type they encounter or secrete in vivo; (4) co-transplantation of hESC-derived endothelial cells and/or cardiac progenitors with hESC-CM results in the formation of functional capillaries; and (5) transplanted hESC-CM survive and mature in vivo for at least 24 weeks. These results form the basis of future developments aiming to reduce the adverse fibrotic reaction that currently complicates cell-based therapies for cardiac disease, and to provide an additional clue towards successful engraftment of cardiomyocytes by co-transplanting endothelial cells.


Asunto(s)
Células Madre Embrionarias/fisiología , Matriz Extracelular/metabolismo , Miocitos Cardíacos/trasplante , Neovascularización Fisiológica , Animales , Diferenciación Celular , Línea Celular , Humanos , Ratones , Miocitos Cardíacos/citología
8.
Cell Mol Life Sci ; 66(5): 800-13, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19151924

RESUMEN

Studies on identification, derivation and characterization of human stem cells in the last decade have led to high expectations in the field of regenerative medicine. Although it is clear that for successful stem cell-based therapy several obstacles have to be overcome, other opportunities lay ahead for the use of human stem cells. A more immediate application would be the development of human models for cell-type specific differentiation and disease in vitro. Cardiomyocytes can be generated from stem cells, which have been shown to follow similar molecular events of cardiac development in vivo. Furthermore, several monogenic cardiovascular diseases have been described, for which in vitro models in stem cells could be generated. Here, we will discuss the potential of human embryonic stem cells, cardiac stem cells and the recently described induced pluripotent stem cells as models for cardiac differentiation and disease.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Corazón , Células Madre Pluripotentes/fisiología , Animales , Cardiomiopatías/fisiopatología , Canalopatías/fisiopatología , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Corazón/anatomía & histología , Corazón/embriología , Humanos , Análisis por Micromatrices , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Células Madre Pluripotentes/citología , Transcripción Genética
9.
Cell Mol Life Sci ; 64(6): 704-18, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17380311

RESUMEN

Investigating the signalling pathways that regulate heart development is essential if stem cells are to become an effective source of cardiomyocytes that can be used for studying cardiac physiology and pharmacology and eventually developing cell-based therapies for heart repair. Here, we briefly describe current understanding of heart development in vertebrates and review the signalling pathways thought to be involved in cardiomyogenesis in multiple species. We discuss how this might be applied to stem cells currently thought to have cardiomyogenic potential by considering the factors relevant for each differentiation step from the undifferentiated cell to nascent mesoderm, cardiac progenitors and finally a fully determined cardiomyocyte. We focus particularly on how this is being applied to human embryonic stem cells and provide recent examples from both our own work and that of others.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Miocitos Cardíacos/citología , Transducción de Señal , Animales , Corazón/embriología , Humanos , Morfogénesis
10.
Handb Exp Pharmacol ; (174): 101-22, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16370326

RESUMEN

Terminal heart failure is characterized by a significant loss of cardiac myocytes. Stem cells represent a possibility for replacing these lost myocytes but the question of which stem cells are most ideally suited for cell transplantation therapies is still being addressed. Here, we consider human embryonic stem cells (HESC), derived from human embryos in this context. We review the methods used to induce their differentiation to cardiomyocytes in culture, their properties in relation to primary human cardiomyocytes and their ability to integrate into host myocardium. In addition, issues regarding their safety that need addressing before use in cell transplantation therapies, both generally and specifically in relation to the heart, are considered.


Asunto(s)
Corazón/embriología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Células Madre/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Perfilación de la Expresión Génica , Humanos
11.
Minerva Cardioangiol ; 52(5): 433-45, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15514577

RESUMEN

The use of stem cells for cardiac repair is a promising opportunity for developing new treatment strategies as the applications are theoretically unlimited and lead to actual cardiac tissue regeneration. Human embryonic stem cells were only recently cloned and their capacity to differentiate into true cardiomyocytes makes them in principle an unlimited source of transplantable cells for cardiac repair, although practical and ethical constraints exist. Also, the study of embryonic stem cells and their differentiation into cardiomyocytes will bring forth new insights into the molecular processes involved in cardiomyocyte-development and -proliferation, which could lead to the development of other strategies to augment in vivo cardiomyocyte numbers. On the other hand, somatic stem cells are alternative cell sources that can be used for cell transplantation purposes. They do not evoke ethical issues and bear less ethical constraints. However, they also appear to be much more restricted in their differentiation potential than the embryonic stem cells. Here we discuss the use of both cell types, embryonic and somatic stem cells, in relation with their importance for the clarification of cardiomyocyte-development and their possible usefulness for clinical therapy.


Asunto(s)
Isquemia Miocárdica/cirugía , Trasplante de Células Madre , Humanos , Isquemia Miocárdica/patología , Miocardio/patología , Miocitos Cardíacos/trasplante , Trasplante de Células Madre/métodos , Resultado del Tratamiento
12.
Neth Heart J ; 12(9): 400-403, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25696371

RESUMEN

Advancements in sequencing technology have made it possible to obtain more information about the DNA sequence, structure and the transcript products of the genome from different species. This information is collected in DNA databases. These databases contain many genes of which the functions have not yet been discovered. By using online biotechnology tools novel genes and their transcripts can be identified. The identification of novel genes using DNA database analysis is referred to as in silico cloning. In silico cloning may not only provide new information on genes and their biological function, it may also lead to identification of molecular targets for drug discovery activities. In this review we describe the process of in silico cloning and its application in biomedical research.

13.
Cardiovasc Res ; 58(2): 423-34, 2003 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-12757876

RESUMEN

AIM: Determination of the phenotype of adult human atrial and ventricular myocytes based on gene expression and morphology. METHODS: Atrial and ventricular cardiomyocytes were obtained from patients undergoing cardiac surgery using a modified isolation procedure. Myocytes were isolated and cultured with or without serum. The relative cell attachment promoting efficiency of several reagents was evaluated and compared. Morphological changes during long-term culture were assessed with phase contrast microscopy, morphometric analysis and immunocytochemistry or RT-PCR of sarcomeric markers including alpha-actinin, myosin light chain-2 (MLC-2) and the adhesion molecule, cadherin. RESULTS: The isolation method produced viable rod-shaped atrial (16.6+/-6.0%, mean+/-S.E.; n=5) and ventricular cells (22.4+/-8.0%, mean+/-S.E.; n=5) in addition to significant numbers of apoptotic and necrotic cells. Cell dedifferentiation was characterized by the loss of sarcomeric structure, condensation and extrusion of sarcomeric proteins. Cells cultured with low serum recovered and assumed a flattened, spread form with two distinct morphologies apparent. Type I cells were large, had extensive sarcolemmal spreading, with stress fibers and nascent myofibrils, whilst type II cells appeared smaller, with more mature myofibril organisation and focal adhesions. CONCLUSION: Characterization of the redifferentiation capabilities of cultured adult cardiac myocytes in culture, provides an important system for comparing cardiomyocytes differentiating from human stem cells and provides the basis for an in vitro transplantation model to study interaction and communication between primary adult and stem cell-derived cardiomyocytes.


Asunto(s)
Contracción Miocárdica/fisiología , Miocitos Cardíacos/fisiología , Miofibrillas/fisiología , Actinina/análisis , Actinas/genética , Adulto , Apoptosis , Factor Natriurético Atrial/genética , Cadherinas/análisis , ATPasas Transportadoras de Calcio/genética , Miosinas Cardíacas/genética , Adhesión Celular/fisiología , Separación Celular/métodos , Células Cultivadas , Expresión Génica , Atrios Cardíacos , Ventrículos Cardíacos , Humanos , Inmunohistoquímica/métodos , Microscopía de Contraste de Fase , Miocitos Cardíacos/citología , Miofibrillas/ultraestructura , Cadenas Ligeras de Miosina/genética , Fenotipo , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcómeros/ultraestructura , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico , Tropomiosina/genética
15.
Dev Biol ; 234(2): 497-509, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11397016

RESUMEN

MEF2C is a MADS-box transcription factor required for cardiac myogenesis and morphogenesis. In MEF2C mutant mouse embryos, heart development arrests at the looping stage (embryonic day 9.0), the future right ventricular chamber fails to form, and cardiomyocyte differentiation is disrupted. To identify genes regulated by MEF2C in the developing heart, we performed differential array analysis coupled with subtractive cloning using RNA from heart tubes of wild-type and MEF2C-null embryos. Here, we describe a novel MEF2C-dependent gene that encodes a cardiac-restricted protein, called CHAMP (cardiac helicase activated by MEF2 protein), that contains seven conserved motifs characteristic of helicases involved in RNA processing, DNA replication, and transcription. During mouse embryogenesis, CHAMP expression commences in the linear heart tube at embryonic day 8.0, shortly after initiation of MEF2C expression in the cardiogenic region. Thereafter, CHAMP is expressed specifically in embryonic and postnatal cardiomyocytes. At the trabeculation stage of heart development, CHAMP expression is highest in the trabecular region in which cardiomyocytes have exited the cell cycle and is lowest in the proliferative compact zone. These findings suggest that CHAMP acts downstream of MEF2C in a cardiac-specific regulatory pathway for RNA processing and/or transcriptional control.


Asunto(s)
ADN Helicasas/genética , Corazón/embriología , Factores Reguladores Miogénicos/metabolismo , ARN Helicasas , Secuencia de Aminoácidos , Animales , Cardiomegalia/etiología , Compartimento Celular , Clonación Molecular/métodos , Regulación hacia Abajo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción MEF2 , Ratones , Ratones Mutantes , Datos de Secuencia Molecular , Especificidad de Órganos , Homología de Secuencia de Aminoácido , Distribución Tisular
16.
J Clin Invest ; 105(10): 1395-406, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10811847

RESUMEN

Hypertrophic growth is an adaptive response of the heart to diverse pathological stimuli and is characterized by cardiomyocyte enlargement, sarcomere assembly, and activation of a fetal program of cardiac gene expression. A variety of Ca(2+)-dependent signal transduction pathways have been implicated in cardiac hypertrophy, but whether these pathways are independent or interdependent and whether there is specificity among them are unclear. Previously, we showed that activation of the Ca(2+)/calmodulin-dependent protein phosphatase calcineurin or its target transcription factor NFAT3 was sufficient to evoke myocardial hypertrophy in vivo. Here, we show that activated Ca(2+)/calmodulin-dependent protein kinases-I and -IV (CaMKI and CaMKIV) also induce hypertrophic responses in cardiomyocytes in vitro and that CaMKIV overexpressing mice develop cardiac hypertrophy with increased left ventricular end-diastolic diameter and decreased fractional shortening. Crossing this transgenic line with mice expressing a constitutively activated form of NFAT3 revealed synergy between these signaling pathways. We further show that CaMKIV activates the transcription factor MEF2 through a posttranslational mechanism in the hypertrophic heart in vivo. Activated calcineurin is a less efficient activator of MEF2-dependent transcription, suggesting that the calcineurin/NFAT and CaMK/MEF2 pathways act in parallel. These findings identify MEF2 as a downstream target for CaMK signaling in the hypertrophic heart and suggest that the CaMK and calcineurin pathways preferentially target different transcription factors to induce cardiac hypertrophy.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares , Factores de Transcripción/metabolismo , Animales , Factor Natriurético Atrial/genética , Calcineurina/metabolismo , Proteína Quinasa Tipo 4 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Cardiomegalia/genética , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Luciferasas/genética , Factores de Transcripción MEF2 , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Factores Reguladores Miogénicos , Cadenas Pesadas de Miosina/genética , Factores de Transcripción NFATC , Regiones Promotoras Genéticas , Ratas , Transducción de Señal
17.
Mech Dev ; 92(2): 277-84, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10727866

RESUMEN

In order to identify novel genes enriched in adult heart, we performed a subtractive hybridization for genes expressed in mouse heart but not in skeletal muscle. We identified two alternative splicing variants of a novel PDZ-LIM domain protein, which we named Oracle. Both variants contain a PDZ domain at the amino-terminus and three LIM domains at the carboxy-terminus. Highest homology of Oracle was found with the human and rat enigma proteins in the PDZ domain (62 and 61%, respectively) and in the LIM domains (60 and 69%, respectively). By Northern hybridization analysis, we showed that expression is highest in adult mouse heart, low in skeletal muscle and undetectable in other adult mouse tissues. In situ hybridization in mouse embryos confirmed and extended these data by showing high expression of Oracle mRNA in atrial and ventricular myocardial cells from E8.5. From E9.5 low expression of Oracle mRNA was detectable in myotomes. These data suggest a role for Oracle in the early development and function of heart and skeletal muscle.


Asunto(s)
Corazón/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Musculares , Músculo Esquelético/fisiología , Empalme Alternativo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Humanos , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , Músculo Esquelético/embriología , Isoformas de Proteínas , ARN Mensajero , Ratas , Homología de Secuencia de Aminoácido
18.
Pharm World Sci ; 20(3): 93-9, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9618731

RESUMEN

Recent developments in our knowledge of the renin-angiotensin system (RAS) necessitate an update of the classical view on this system. These developments pertain to the pathways leading to formation of angiotensin II and other active metabolites, their receptors, biological functions and the presence of renin-angiotensin systems in tissues. The implications of the above new developments for the current interest in tissue renin-angiotensin systems as potential targets for drug therapy in cardiovascular disease are discussed in this review.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Angiotensina II/biosíntesis , Inhibidores de la Enzima Convertidora de Angiotensina/química , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Peptidil-Dipeptidasa A/genética , Polimorfismo Genético/efectos de los fármacos , Receptores de Angiotensina/química , Receptores de Angiotensina/fisiología , Sistema Renina-Angiotensina/fisiología
19.
J Mater Sci Mater Med ; 9(12): 749-54, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15348934

RESUMEN

Tissue engineering of articular cartilage in order to restore the function of degenerated, diarthrodial joints is currently widely under investigation. The results obtained thus far indicate that proper control of the differentiation of the cells used for this purpose is essential to produce and maintain a hyaline-like matrix. In this study, a procedure is described by which differentiation of chondrocytes in vitro and ex vivo can be studied. The method involves quantitative assessment of mRNA for different collagens, which are markers for differentiation of chondrocytes, by competitive PCR. In a culture system employing human osteoarthritic chondrocytes, mRNAs for the alpha1-chains of collagen types I, II and X are quantified. The procedure is fast, specific and sensitive. However, several controls should be included to ascertain the reliability of the assessment.

20.
Am J Physiol ; 271(3 Pt 2): H1040-8, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8853339

RESUMEN

Wistar-Kyoto rats underwent myocardial infarction (MI) or sham surgery. At different time points after surgery (1-90 days), hearts were removed and divided into infarcted left ventricle (LV), noninfarcted septum, and right ventricle. The tissues were used for total RNA isolation or Formalin fixation for in situ hybridization (ISH). Renin and angiotensinogen mRNA contents were quantified by the competitive reverse transcriptase polymerase chain reaction. We found a 4-, 14-, and 8-fold increase (P < 0.05, n = 6) in renin mRNA in the infarcted LV at 2, 4, and 7 days after MI, respectively. No differences were observed between angiotensinogen mRNA levels in sham and infarcted hearts. ISH at 4 days after surgery revealed a dense renin mRNA labeling around the infarcted area, whereas ISH of angiotensinogen displayed an overall low density in the myocardium with somewhat higher levels in the epicardium of sham and MI animals. Atrial natriuretic factor mRNA, a marker for cardiac hypertrophy, was approximately twofold higher in all compartments of the hearts after MI. The low amounts of renin and angiotensinogen mRNA in the noninfarcted hypertrophied myocardium indicate that the intracardiac synthesis of these components does not play a dominant role in the development of cardiac hypertrophy in the rat heart after MI. In addition, the increased renin mRNA expression in the border zone of the infarcted LV suggests a role for intracardiac angiotensin II in infarct healing.


Asunto(s)
Angiotensinógeno/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Renina/metabolismo , Angiotensinógeno/genética , Animales , Factor Natriurético Atrial/genética , Hibridación in Situ , Masculino , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas WKY , Renina/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA