Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Neurophysiol ; 117(3): 1320-1341, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031399

RESUMEN

Brain in vitro models are critically important to developing our understanding of basic nervous system cellular physiology, potential neurotoxic effects of chemicals, and specific cellular mechanisms of many disease states. In this study, we sought to address key shortcomings of current brain in vitro models: the scarcity of comparative data for cells originating from distinct brain regions and the lack of multiregional brain in vitro models. We demonstrated that rat neurons from different brain regions exhibit unique profiles regarding their cell composition, protein expression, metabolism, and electrical activity in vitro. In vivo, the brain is unique in its structural and functional organization, and the interactions and communication between different brain areas are essential components of proper brain function. This fact and the observation that neurons from different areas of the brain exhibit unique behaviors in vitro underline the importance of establishing multiregional brain in vitro models. Therefore, we here developed a multiregional brain-on-a-chip and observed a reduction of overall firing activity, as well as altered amounts of astrocytes and specific neuronal cell types compared with separately cultured neurons. Furthermore, this multiregional model was used to study the effects of phencyclidine, a drug known to induce schizophrenia-like symptoms in vivo, on individual brain areas separately while monitoring downstream effects on interconnected regions. Overall, this work provides a comparison of cells from different brain regions in vitro and introduces a multiregional brain-on-a-chip that enables the development of unique disease models incorporating essential in vivo features.NEW & NOTEWORTHY Due to the scarcity of comparative data for cells from different brain regions in vitro, we demonstrated that neurons isolated from distinct brain areas exhibit unique behaviors in vitro. Moreover, in vivo proper brain function is dependent on the connection and communication of several brain regions, underlining the importance of developing multiregional brain in vitro models. We introduced a novel brain-on-a-chip model, implementing essential in vivo features, such as different brain areas and their functional connections.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/citología , Neuronas/clasificación , Neuronas/fisiología , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Células Cultivadas , Cromatografía Líquida de Alta Presión , Femenino , Expresión Génica/fisiología , Glutamato Descarboxilasa/metabolismo , Alucinógenos/farmacología , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Consumo de Oxígeno , Fenciclidina/farmacología , Análisis de Componente Principal , Mapas de Interacción de Proteínas , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
2.
Proc Natl Acad Sci U S A ; 110(24): 9770-5, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23716679

RESUMEN

The lack of a robust pipeline of medical therapeutic agents for the treatment of heart disease may be partially attributed to the lack of in vitro models that recapitulate the essential structure-function relationships of healthy and diseased myocardium. We designed and built a system to mimic mechanical overload in vitro by applying cyclic stretch to engineered laminar ventricular tissue on a stretchable chip. To test our model, we quantified changes in gene expression, myocyte architecture, calcium handling, and contractile function and compared our results vs. several decades of animal studies and clinical observations. Cyclic stretch activated gene expression profiles characteristic of pathological remodeling, including decreased α- to ß-myosin heavy chain ratios, and induced maladaptive changes to myocyte shape and sarcomere alignment. In stretched tissues, calcium transients resembled those reported in failing myocytes and peak systolic stress was significantly reduced. Our results suggest that failing myocardium, as defined genetically, structurally, and functionally, can be replicated in an in vitro microsystem by faithfully recapitulating the structural and mechanical microenvironment of the diseased heart.


Asunto(s)
Insuficiencia Cardíaca/genética , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Remodelación Ventricular/genética , Animales , Animales Recién Nacidos , Calcio/metabolismo , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos , Modelos Cardiovasculares , Contracción Miocárdica/genética , Miocardio/patología , Cadenas Pesadas de Miosina/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas Sprague-Dawley , Sarcómeros/metabolismo , Sístole/genética , Factores de Tiempo , Miosinas Ventriculares/genética
3.
Am J Pathol ; 181(6): 2030-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23159216

RESUMEN

The heart actively remodels architecture in response to various physiological and pathological conditions. Gross structural change of the heart chambers is directly reflected at the cellular level by altering the morphological characteristics of individual cardiomyocytes. However, an understanding of the relationship between cardiomyocyte shape and the contractile function remains unclear. By using in vitro assays to analyze systolic stress of cardiomyocytes with controlled shape, we demonstrated that the characteristic morphological features of cardiomyocytes observed in a variety of pathophysiological conditions are correlated with mechanical performance. We found that cardiomyocyte contractility is optimized at the cell length/width ratio observed in normal hearts, and decreases in cardiomyocytes with morphological characteristics resembling those isolated from failing hearts. Quantitative analysis of sarcomeric architecture revealed that the change of contractility may arise from alteration of myofibrillar structure. Measurements of intracellular calcium in myocytes revealed unique characteristics of calcium metabolism as a function of myocyte shape. Our data suggest that cell shape is critical in determining contractile performance of single cardiomyocytes by regulating the intracellular structure and calcium handling ability.


Asunto(s)
Forma de la Célula , Procesamiento de Imagen Asistido por Computador , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Sarcómeros/fisiología , Animales , Calcio/metabolismo , ADN/metabolismo , Diástole/fisiología , Ratas , Ratas Sprague-Dawley , Sístole/fisiología
4.
PLoS Comput Biol ; 7(2): e1001088, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21390276

RESUMEN

The organization of muscle is the product of functional adaptation over several length scales spanning from the sarcomere to the muscle bundle. One possible strategy for solving this multiscale coupling problem is to physically constrain the muscle cells in microenvironments that potentiate the organization of their intracellular space. We hypothesized that boundary conditions in the extracellular space potentiate the organization of cytoskeletal scaffolds for directed sarcomeregenesis. We developed a quantitative model of how the cytoskeleton of neonatal rat ventricular myocytes organizes with respect to geometric cues in the extracellular matrix. Numerical results and in vitro assays to control myocyte shape indicated that distinct cytoskeletal architectures arise from two temporally-ordered, organizational processes: the interaction between actin fibers, premyofibrils and focal adhesions, as well as cooperative alignment and parallel bundling of nascent myofibrils. Our results suggest that a hierarchy of mechanisms regulate the self-organization of the contractile cytoskeleton and that a positive feedback loop is responsible for initiating the break in symmetry, potentiated by extracellular boundary conditions, is required to polarize the contractile cytoskeleton.


Asunto(s)
Modelos Biológicos , Miocitos Cardíacos/fisiología , Miofibrillas/fisiología , Actinas/metabolismo , Animales , Células Cultivadas , Simulación por Computador , Citoesqueleto/metabolismo , Citoesqueleto/fisiología , Adhesiones Focales/química , Adhesiones Focales/fisiología , Inmunohistoquímica , Contracción Muscular/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miofibrillas/química , Miofibrillas/metabolismo , Ratas , Ratas Sprague-Dawley , Sarcómeros/metabolismo , Sarcómeros/fisiología
5.
Adv Biosyst ; 4(9): e1900230, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32744807

RESUMEN

The functional state of the neurovascular unit (NVU), composed of the blood-brain barrier and the perivasculature that forms a dynamic interface between the blood and the central nervous system (CNS), plays a central role in the control of brain homeostasis and is strongly affected by CNS drugs. Human primary brain microvascular endothelium, astrocyte, pericyte, and neural cell cultures are often used to study NVU barrier functions as well as drug transport and efficacy; however, the proteomic and metabolomic responses of these different cell types are not well characterized. Culturing each cell type separately, using deep coverage proteomic analysis and characterization of the secreted metabolome, as well as measurements of mitochondrial activity, the responses of these cells under baseline conditions and when exposed to the NVU-impairing stimulant methamphetamine (Meth) are analyzed. These studies define the previously unknown metabolic and proteomic profiles of human brain pericytes and lead to improved characterization of the phenotype of each of the NVU cell types as well as cell-specific metabolic and proteomic responses to Meth.


Asunto(s)
Metaboloma/efectos de los fármacos , Metanfetamina/farmacología , Neuronas , Pericitos , Proteoma/efectos de los fármacos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Encéfalo/irrigación sanguínea , Encéfalo/citología , Células Cultivadas , Estimulantes del Sistema Nervioso Central/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Humanos , Metabolómica , Neuronas/citología , Neuronas/efectos de los fármacos , Pericitos/citología , Pericitos/efectos de los fármacos , Proteoma/análisis , Proteómica
6.
Sci Transl Med ; 12(565)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33055246

RESUMEN

Extracellular vesicles (EVs) derived from various stem cell sources induce cardioprotective effects during ischemia-reperfusion injury (IRI). These have been attributed mainly to the antiapoptotic, proangiogenic, microRNA (miRNA) cargo within the stem cell-derived EVs. However, the mechanisms of EV-mediated endothelial signaling to cardiomyocytes, as well as their therapeutic potential toward ischemic myocardial injury, are not clear. EV content beyond miRNA that may contribute to cardioprotection has not been fully illuminated. This study characterized the protein cargo of human vascular endothelial EVs (EEVs) to identify lead cardioactive proteins and assessed the effect of EEVs on human laminar cardiac tissues (hlCTs) exposed to IRI. We mapped the protein content of human vascular EEVs and identified proteins that were previously associated with cellular metabolism, redox state, and calcium handling, among other processes. Analysis of the protein landscape of human cardiomyocytes revealed corresponding modifications induced by EEV treatment. To assess their human-specific cardioprotection in vitro, we developed a human heart-on-a-chip IRI assay using human stem cell-derived, engineered cardiac tissues. We found that EEVs alleviated cardiac cell death as well as the loss in contractile capacity during and after simulated IRI in an uptake- and dose-dependent manner. Moreover, we found that EEVs increased the respiratory capacity of normoxic cardiomyocytes. These results suggest that vascular EEVs rescue hlCTs exposed to IRI possibly by supplementing injured myocytes with cargo that supports multiple metabolic and salvage pathways and therefore may serve as a multitargeted therapy for IRI.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Daño por Reperfusión , Apoptosis , Humanos , Miocitos Cardíacos
7.
Cell Motil Cytoskeleton ; 65(8): 641-51, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18561184

RESUMEN

Cardiac organogenesis and pathogenesis are both characterized by changes in myocyte shape, cytoskeletal architecture, and the extracellular matrix (ECM). However, the mechanisms by which the ECM influences myocyte shape and myofibrillar patterning are unknown. We hypothesized that geometric cues in the ECM align sarcomeres by directing the actin network orientation. To test our hypothesis, we cultured neonatal rat ventricular myocytes on islands of micro-patterned ECM to measure how they remodeled their cytoskeleton in response to extracellular cues. Myocytes spread and assumed the shape of circular and rectangular islands and reorganized their cytoskeletons and myofibrillar arrays with respect to the ECM boundary conditions. Circular myocytes did not assemble predictable actin networks nor organized sarcomere arrays. In contrast, myocytes cultured on rectangular ECM patterns with aspect ratios ranging from 1:1 to 7:1 aligned their sarcomeres in predictable and repeatable patterns based on highly localized focal adhesion complexes. Examination of averaged alpha-actinin images revealed invariant sarcomeric registration irrespective of myocyte aspect ratio. Since the sarcomere sub-units possess a fixed length, this observation indicates that cytoskeleton configuration is length-limited by the extracellular boundary conditions. These results indicate that modification of the extracellular microenvironment induces dynamic reconfiguring of the myocyte shape and intracellular architecture. Furthermore, geometric boundaries such as corners induce localized myofibrillar anisotropy that becomes global as the myocyte aspect ratio increases.


Asunto(s)
Forma de la Célula/fisiología , Células Musculares/metabolismo , Sarcómeros/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Microscopía Fluorescente , Células Musculares/citología , Miofibrillas/metabolismo , Ratas , Ratas Sprague-Dawley , Vinculina/metabolismo
8.
PLoS One ; 13(3): e0194706, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29590169

RESUMEN

Cardiac tissue development and pathology have been shown to depend sensitively on microenvironmental mechanical factors, such as extracellular matrix stiffness, in both in vivo and in vitro systems. We present a novel quantitative approach to assess cardiac structure and function by extending the classical traction force microscopy technique to tissue-level preparations. Using this system, we investigated the relationship between contractile proficiency and metabolism in neonate rat ventricular myocytes (NRVM) cultured on gels with stiffness mimicking soft immature (1 kPa), normal healthy (13 kPa), and stiff diseased (90 kPa) cardiac microenvironments. We found that tissues engineered on the softest gels generated the least amount of stress and had the smallest work output. Conversely, cardiomyocytes in tissues engineered on healthy- and disease-mimicking gels generated significantly higher stresses, with the maximal contractile work measured in NRVM engineered on gels of normal stiffness. Interestingly, although tissues on soft gels exhibited poor stress generation and work production, their basal metabolic respiration rate was significantly more elevated than in other groups, suggesting a highly ineffective coupling between energy production and contractile work output. Our novel platform can thus be utilized to quantitatively assess the mechanotransduction pathways that initiate tissue-level structural and functional remodeling in response to substrate stiffness.


Asunto(s)
Mecanotransducción Celular , Microscopía de Fuerza Atómica/métodos , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Estrés Mecánico , Ingeniería de Tejidos/métodos , Animales , Animales Recién Nacidos , Células Cultivadas , Ratas , Ratas Sprague-Dawley
9.
Biofabrication ; 10(2): 025004, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29337695

RESUMEN

Organ-on-chip platforms aim to improve preclinical models for organ-level responses to novel drug compounds. Heart-on-a-chip assays in particular require tissue engineering techniques that rely on labor-intensive photolithographic fabrication or resolution-limited 3D printing of micropatterned substrates, which limits turnover and flexibility of prototyping. We present a rapid and automated method for large scale on-demand micropatterning of gelatin hydrogels for organ-on-chip applications using a novel biocompatible laser-etching approach. Fast and automated micropatterning is achieved via photosensitization of gelatin using riboflavin-5'phosphate followed by UV laser-mediated photoablation of the gel surface in user-defined patterns only limited by the resolution of the 15 µm wide laser focal point. Using this photopatterning approach, we generated microscale surface groove and pillar structures with feature dimensions on the order of 10-30 µm. The standard deviation of feature height was 0.3 µm, demonstrating robustness and reproducibility. Importantly, the UV-patterning process is non-destructive and does not alter gelatin micromechanical properties. Furthermore, as a quality control step, UV-patterned heart chip substrates were seeded with rat or human cardiac myocytes, and we verified that the resulting cardiac tissues achieved structural organization, contractile function, and long-term viability comparable to manually patterned gelatin substrates. Start-to-finish, UV-patterning shortened the time required to design and manufacture micropatterned gelatin substrates for heart-on-chip applications by up to 60% compared to traditional lithography-based approaches, providing an important technological advance enroute to automated and continuous manufacturing of organ-on-chips.


Asunto(s)
Hidrogeles/química , Análisis de Matrices Tisulares/instrumentación , Ingeniería de Tejidos/instrumentación , Andamios del Tejido/química , Animales , Automatización , Células Cultivadas , Gelatina/química , Humanos , Miocitos Cardíacos/citología , Impresión Tridimensional , Ratas
10.
Biomaterials ; 166: 96-108, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29549768

RESUMEN

Wounds in the fetus can heal without scarring. Consequently, biomaterials that attempt to recapitulate the biophysical and biochemical properties of fetal skin have emerged as promising pro-regenerative strategies. The extracellular matrix (ECM) protein fibronectin (Fn) in particular is believed to play a crucial role in directing this regenerative phenotype. Accordingly, Fn has been implicated in numerous wound healing studies, yet remains untested in its fibrillar conformation as found in fetal skin. Here, we show that high extensional (∼1.2 ×105 s-1) and shear (∼3 ×105 s-1) strain rates in rotary jet spinning (RJS) can drive high throughput Fn fibrillogenesis (∼10 mL/min), thus producing nanofiber scaffolds that are used to effectively enhance wound healing. When tested on a full-thickness wound mouse model, Fn nanofiber dressings not only accelerated wound closure, but also significantly improved tissue restoration, recovering dermal and epidermal structures as well as skin appendages and adipose tissue. Together, these results suggest that bioprotein nanofiber fabrication via RJS could set a new paradigm for enhancing wound healing and may thus find use in a variety of regenerative medicine applications.


Asunto(s)
Materiales Biocompatibles , Fibronectinas , Nanofibras , Cicatrización de Heridas , Administración Cutánea , Animales , Materiales Biocompatibles/química , Fibronectinas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Nanofibras/química , Piel/efectos de los fármacos , Piel/patología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA