Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Banco de datos
Tipo de estudio
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biomech Eng ; 144(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34382649

RESUMEN

Through a variety of mechanisms, a healthy heart is able to regulate its structure and dynamics across multiple length scales. Disruption of these mechanisms can have a cascading effect, resulting in severe structural and/or functional changes that permeate across different length scales. Due to this hierarchical structure, there is interest in understanding how the components at the various scales coordinate and influence each other. However, much is unknown regarding how myofibril bundles are organized within a densely packed cell and the influence of the subcellular components on the architecture that is formed. To elucidate potential factors influencing cytoskeletal development, we proposed a computational model that integrated interactions at both the cellular and subcellular scale to predict the location of individual myofibril bundles that contributed to the formation of an energetically favorable cytoskeletal network. Our model was tested and validated using experimental metrics derived from analyzing single-cell cardiomyocytes. We demonstrated that our model-generated networks were capable of reproducing the variation observed in experimental cells at different length scales as a result of the stochasticity inherent in the different interactions between the various cellular components. Additionally, we showed that incorporating length-scale parameters resulted in physical constraints that directed cytoskeletal architecture toward a structurally consistent motif. Understanding the mechanisms guiding the formation and organization of the cytoskeleton in individual cardiomyocytes can aid tissue engineers toward developing functional cardiac tissue.


Asunto(s)
Citoesqueleto , Miocitos Cardíacos , Microtúbulos , Miocitos Cardíacos/fisiología , Miofibrillas , Ingeniería de Tejidos
2.
Ann Biomed Eng ; 49(12): 3524-3539, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34585335

RESUMEN

Genetic mutations to the Lamin A/C gene (LMNA) can cause heart disease, but the mechanisms making cardiac tissues uniquely vulnerable to the mutations remain largely unknown. Further, patients with LMNA mutations have highly variable presentation of heart disease progression and type. In vitro patient-specific experiments could provide a powerful platform for studying this phenomenon, but the use of induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) introduces heterogeneity in maturity and function thus complicating the interpretation of the results of any single experiment. We hypothesized that integrating single cell RNA sequencing (scRNA-seq) with analysis of the tissue architecture and contractile function would elucidate some of the probable mechanisms. To test this, we investigated five iPSC-CM lines, three controls and two patients with a (c.357-2A>G) mutation. The patient iPSC-CM tissues had significantly weaker stress generation potential than control iPSC-CM tissues demonstrating the viability of our in vitro approach. Through scRNA-seq, differentially expressed genes between control and patient lines were identified. Some of these genes, linked to quantitative structural and functional changes, were cardiac specific, explaining the targeted nature of the disease progression seen in patients. The results of this work demonstrate the utility of combining in vitro tools in exploring heart disease mechanics.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Expresión Génica , Células Madre Pluripotentes Inducidas/citología , Lamina Tipo A/genética , Contracción Miocárdica , Miocitos Cardíacos/fisiología , Adulto , Anciano , Línea Celular , Humanos , Persona de Mediana Edad
3.
J Vis Exp ; (153)2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31814616

RESUMEN

Science relies on increasingly complex data sets for progress, but common data management methods such as spreadsheet programs are inadequate for the growing scale and complexity of this information. While database management systems have the potential to rectify these issues, they are not commonly utilized outside of business and informatics fields. Yet, many research labs already generate "medium sized", low velocity, multi-dimensional data that could greatly benefit from implementing similar systems. In this article, we provide a conceptual overview explaining how databases function and the advantages they provide in tissue engineering applications. Structural fibroblast data from individuals with a lamin A/C mutation was used to illustrate examples within a specific experimental context. Examples include visualizing multidimensional data, linking tables in a relational database structure, mapping a semi-automated data pipeline to convert raw data into structured formats, and explaining the underlying syntax of a query. Outcomes from analyzing the data were used to create plots of various arrangements and significance was demonstrated in cell organization in aligned environments between the positive control of Hutchinson-Gilford progeria, a well-known laminopathy, and all other experimental groups. In comparison to spreadsheets, database methods were enormously time efficient, simple to use once set up, allowed for immediate access of original file locations, and increased data rigor. In response to the National Institutes of Health (NIH) emphasis on experimental rigor, it is likely that many scientific fields will eventually adopt databases as common practice due to their strong capability to effectively organize complex data.


Asunto(s)
Sistemas de Administración de Bases de Datos , Bases de Datos Factuales , Ingeniería de Tejidos , Línea Celular , Humanos , Lamina Tipo A/genética , Estados Unidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA