Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38904851

RESUMEN

Computational, or in-silico, models are an effective, non-invasive tool for investigating cardiovascular function. These models can be used in the analysis of experimental and clinical data to identify possible mechanisms of (ab)normal cardiovascular physiology. Recent advances in computing power and data management have led to innovative and complex modeling frameworks that simulate cardiovascular function across multiple scales. While commonly used in multiple disciplines, there is a lack of concise guidelines for the implementation of computer models in cardiovascular research. In line with recent calls for more reproducible research, it is imperative that scientists adhere to credible practices when developing and applying computational models to their research. The goal of this manuscript is to provide a consensus document that identifies best practices for in-silico computational modeling in cardiovascular research. These guidelines provide the necessary methods for mechanistic model development, model analysis, and formal model calibration using fundamentals from statistics. We outline rigorous practices for computational modeling in cardiovascular research and discuss its synergistic value to experimental and clinical data.

2.
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
3.
PLoS Comput Biol ; 16(3): e1007676, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130207

RESUMEN

As sarcomeres produce the force necessary for contraction, assessment of sarcomere order is paramount in evaluation of cardiac and skeletal myocytes. The uniaxial force produced by sarcomeres is ideally perpendicular to their z-lines, which couple parallel myofibrils and give cardiac and skeletal myocytes their distinct striated appearance. Accordingly, sarcomere structure is often evaluated by staining for z-line proteins such as α-actinin. However, due to limitations of current analysis methods, which require manual or semi-manual handling of images, the mechanism by which sarcomere and by extension z-line architecture can impact contraction and which characteristics of z-line architecture should be used to assess striated myocytes has not been fully explored. Challenges such as isolating z-lines from regions of off-target staining that occur along immature stress fibers and cell boundaries and choosing metrics to summarize overall z-line architecture have gone largely unaddressed in previous work. While an expert can qualitatively appraise tissues, these challenges leave researchers without robust, repeatable tools to assess z-line architecture across different labs and experiments. Additionally, the criteria used by experts to evaluate sarcomeric architecture have not been well-defined. We address these challenges by providing metrics that summarize different aspects of z-line architecture that correspond to expert tissue quality assessment and demonstrate their efficacy through an examination of engineered tissues and single cells. In doing so, we have elucidated a mechanism by which highly elongated cardiomyocytes become inefficient at producing force. Unlike previous manual or semi-manual methods, characterization of z-line architecture using the metrics discussed and implemented in this work can quantitatively evaluate engineered tissues and contribute to a robust understanding of the development and mechanics of striated muscles.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Fibras Musculares Esqueléticas , Miocitos Cardíacos , Sarcómeros , Algoritmos , Animales , Células Cultivadas , Humanos , Microscopía Fluorescente , Fibras Musculares Esqueléticas/química , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/ultraestructura , Miofibrillas/fisiología , Ratas , Ratas Sprague-Dawley , Sarcómeros/química , Sarcómeros/ultraestructura
4.
BMC Med Genet ; 21(1): 152, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32698886

RESUMEN

BACKGROUND: Intermediate filament proteins that construct the nuclear lamina of a cell include the Lamin A/C proteins encoded by the LMNA gene, and are implicated in fundamental processes such as nuclear structure, gene expression, and signal transduction. LMNA mutations predominantly affect mesoderm-derived cell lineages in diseases collectively termed as laminopathies that include dilated cardiomyopathy with conduction defects, different forms of muscular dystrophies, and premature aging syndromes as Hutchinson-Gilford Progeria Syndrome. At present, our understanding of the molecular mechanisms regulating tissue-specific manifestations of laminopathies are still limited. METHODS: To gain deeper insight into the molecular mechanism of a novel LMNA splice-site mutation (c.357-2A > G) in an affected family with cardiac disease, we conducted deep RNA sequencing and pathway analysis for nine fibroblast samples obtained from three patients with cardiomyopathy, three unaffected family members, and three unrelated, unaffected individuals. We validated our findings by quantitative PCR and protein studies. RESULTS: We identified eight significantly differentially expressed genes between the mutant and non-mutant fibroblasts, that included downregulated insulin growth factor binding factor protein 5 (IGFBP5) in patient samples. Pathway analysis showed involvement of the ERK/MAPK signaling pathway consistent with previous studies. We found no significant differences in gene expression for Lamin A/C and B-type lamins between the groups. In mutant fibroblasts, RNA-seq confirmed that only the LMNA wild type allele predominately was expressed, and Western Blot showed normal Lamin A/C protein levels. CONCLUSIONS: IGFBP5 may contribute in maintaining signaling pathway homeostasis, which may lead to the absence of notable molecular and structural abnormalities in unaffected tissues such as fibroblasts. Compensatory mechanisms from other nuclear membrane proteins were not found. Our results also demonstrate that only one copy of the wild type allele is sufficient for normal levels of Lamin A/C protein to maintain physiological function in an unaffected cell type. This suggests that affected cell types such as cardiac tissues may be more sensitive to haploinsufficiency of Lamin A/C. These results provide insight into the molecular mechanism of disease with a possible explanation for the tissue specificity of LMNA-related dilated cardiomyopathy.


Asunto(s)
Cardiomiopatías/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Lamina Tipo A/genética , Transducción de Señal/genética , Secuencia de Bases , Familia , Regulación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/genética , Lámina Nuclear/metabolismo
5.
J Biomech Eng ; 142(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31233093

RESUMEN

Although mutations in the Lamin A/C gene (LMNA) cause a variety of devastating diseases, the pathological mechanism is often unknown. Lamin A/C proteins play a crucial role in forming a meshwork under the nuclear membrane, providing the nucleus with mechanical integrity and interacting with other proteins for gene regulation. Most LMNA mutations result in heart diseases, including some types that primarily have heart disease as the main pathology. In this study, we used cells from patients with different LMNA mutations that primarily lead to heart disease. Indeed, it is a mystery why a mutation to the protein in every nucleus of the body manifests as a disease of primarily the heart in these patients. Here, we aimed to investigate if strains mimicking those within the myocardial environment are sufficient to cause differences in cells with and without the LMNA mutation. To test this, a stretcher device was used to induce cyclic strain upon cells, and viability/proliferation, cytoskeleton and extracellular matrix organization, and nuclear morphology were quantified. The properties of cells with Hutchinson-Gilford progeria syndrome (HGPS) were found to be significantly different from all other cell lines and were mostly in line with previous findings. However, the properties of cells from patients who primarily had heart diseases were not drastically different when compared to individuals without the LMNA mutation. Our results indicated that cyclic strain alone was insufficient to cause any significant differences that could explain the mechanisms that lead to heart diseases in these patients with LMNA mutations.


Asunto(s)
Lamina Tipo A , Progeria , Núcleo Celular , Fibroblastos , Regulación de la Expresión Génica , Humanos , Mutación
6.
J Biomech Eng ; 140(8)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30003248

RESUMEN

Mechanical cues including stretch, compression, and shear stress play a critical role in regulating the behavior of many cell types, particularly those that experience substantial mechanical stress within tissues. Devices that impart mechanical stimulation to cells in vitro have been instrumental in helping to develop a better understanding of how cells respond to mechanical forces. However, these devices often have constraints, such as cost and limited functional capabilities, that restrict their use in research or educational environments. Here, we describe a low-cost method to fabricate a uniaxial cell stretcher that would enable widespread use and facilitate engineering design and mechanobiology education for undergraduate students. The device is capable of producing consistent and reliable strain profiles through the use of a servomotor, gear, and gear rack system. The servomotor can be programmed to output various waveforms at specific frequencies and stretch amplitudes by controlling the degree of rotation, speed, and acceleration of the servogear. In addition, the stretchable membranes are easy to fabricate and can be customized, allowing for greater flexibility in culture well size. We used the custom-built stretching device to uniaxially strain macrophages and cardiomyocytes, and found that both cell types displayed functional and cell shape changes that were consistent with the previous studies using commercially available systems. Overall, this uniaxial cell stretcher provides a more cost-effective alternative to study the effects of mechanical stretch on cells, and can therefore, be widely used in research and educational environments to broaden the study and pedagogy of cell mechanobiology.


Asunto(s)
Biofisica/educación , Células , Costos y Análisis de Costo , Estrés Mecánico , Enseñanza , Animales , Fenómenos Biomecánicos , Ratas
7.
Biophys J ; 110(7): 1615-1624, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27074686

RESUMEN

The heart is a complex organ whose structure and function are intricately linked at multiple length scales. Although several advancements have been achieved in the field of cardiac tissue engineering, current in vitro cardiac tissues do not fully replicate the structure or function necessary for effective cardiac therapy and cardiotoxicity studies. This is partially due to a deficiency in current understandings of cardiac tissue organization's potential downstream effects, such as changes in gene expression levels. We developed a novel (to our knowledge) in vitro tool that can be used to decouple and quantify the contribution of organization and associated downstream effects to tissue function. To do so, cardiac tissue monolayers were designed into a parquet pattern to be organized anisotropically on a local scale, within a parquet tile, and with any desired organization on a global scale. We hypothesized that if the downstream effects were muted, the relationship between developed force and tissue organization could be modeled as a sum of force vectors. With the in vitro experimental platforms of parquet tissues and heart-on-a-chip devices, we were able to prove this hypothesis for both systolic and diastolic stresses. Thus, insight was gained into the relationship between the generated stress and global myofibril organization. Furthermore, it was demonstrated that the developed quantitative tool could be used to estimate the changes in stress production due to downstream effects decoupled from tissue architecture. This has the potential to elucidate properties coupled to tissue architecture, which change force production and pumping function in the diseased heart or stem cell-derived tissues.


Asunto(s)
Fenómenos Mecánicos , Contracción Miocárdica , Miocardio/citología , Animales , Fenómenos Biomecánicos , Ratas
8.
PLoS Comput Biol ; 11(4): e1004190, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25849553

RESUMEN

In biology, organization at multiple scales potentiates biological function. Current advances in staining and imaging of biological tissues provide a wealth of data, but there are few metrics to quantitatively describe these findings. In particular there is a need for a metric that would characterize the correlation and consistency of orientation of different biological constructs within a tissue. We aimed to create such a metric and to demonstrate its use with images of cardiac tissues. The co-orientational order parameter (COOP) was based on the mathematical framework of a classical parameter, the orientational order parameter (OOP). Theorems were proven to illustrate the properties and boundaries of the COOP, which was then applied to both synthetic and experimental data. We showed the COOP to be useful for quantifying the correlation of orientation of constructs such as actin filaments and sarcomeric Z-lines. As expected, cardiac tissues showed perfect correlation between actin filaments and Z-lines. We also demonstrated the use of COOP to quantify the consistency of construct orientation within cells of the same shape. The COOP provides a quantitative tool to characterize tissues beyond co-localization or single construct orientation distribution. In the future, this new parameter could be used to represent the quantitative changes during maturation of cardiac tissue, pathological malformation, and other processes.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Biología Computacional/métodos , Citoesqueleto/ultraestructura , Procesamiento de Imagen Asistido por Computador/métodos , Animales , Miocitos Cardíacos/química , Miocitos Cardíacos/citología , Ratas
9.
J Biomech Eng ; 138(11)2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27617880

RESUMEN

In a properly contracting cardiac muscle, many different subcellular structures are organized into an intricate architecture. While it has been observed that this organization is altered in pathological conditions, the relationship between length-scales and architecture has not been properly explored. In this work, we utilize a variety of architecture metrics to quantify organization and consistency of single structures over multiple scales, from subcellular to tissue scale as well as correlation of organization of multiple structures. Specifically, as the best way to characterize cardiac tissues, we chose the orientational and co-orientational order parameters (COOPs). Similarly, neonatal rat ventricular myocytes were selected for their consistent architectural behavior. The engineered cells and tissues were stained for four architectural structures: actin, tubulin, sarcomeric z-lines, and nuclei. We applied the orientational metrics to cardiac cells of various shapes, isotropic cardiac tissues, and anisotropic globally aligned tissues. With these novel tools, we discovered: (1) the relationship between cellular shape and consistency of self-assembly; (2) the length-scales at which unguided tissues self-organize; and (3) the correlation or lack thereof between organization of actin fibrils, sarcomeric z-lines, tubulin fibrils, and nuclei. All of these together elucidate some of the current mysteries in the relationship between force production and architecture, while raising more questions about the effect of guidance cues on self-assembly function. These types of metrics are the future of quantitative tissue engineering in cardiovascular biomechanics.


Asunto(s)
Ventrículos Cardíacos/citología , Ventrículos Cardíacos/crecimiento & desarrollo , Contracción Miocárdica/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Ingeniería de Tejidos/instrumentación , Animales , Animales Recién Nacidos , Polaridad Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Diseño de Equipo , Análisis de Falla de Equipo , Ratas , Ratas Sprague-Dawley , Ingeniería de Tejidos/métodos , Andamios del Tejido
10.
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
11.
Proc Natl Acad Sci U S A ; 108(50): 19943-8, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22123981

RESUMEN

Endothelial-mesenchymal transformation (EMT) is a critical event for the embryonic morphogenesis of cardiac valves. Inducers of EMT during valvulogenesis include VEGF, TGF-ß1, and wnt/ß-catenin (where wnt refers to the wingless-type mammary tumor virus integration site family of proteins), that are regulated in a spatiotemporal manner. EMT has also been observed in diseased, strain-overloaded valve leaflets, suggesting a regulatory role for mechanical strain. Although the preponderance of studies have focused on the role of soluble mitogens, we asked if the valve tissue microenvironment contributed to EMT. To recapitulate these microenvironments in a controlled, in vitro environment, we engineered 2D valve endothelium from sheep valve endothelial cells, using microcontact printing to mimic the regions of isotropy and anisotropy of the leaflet, and applied cyclic mechanical strain in an attempt to induce EMT. We measured EMT in response to both low (10%) and high strain (20%), where low-strain EMT occurred via increased TGF-ß1 signaling and high strain via increased wnt/ß-catenin signaling, suggesting dual strain-dependent routes to distinguish EMT in healthy versus diseased valve tissue. The effect was also directionally dependent, where cyclic strain applied orthogonal to axis of the engineered valve endothelium alignment resulted in severe disruption of cell microarchitecture and greater EMT. Once transformed, these tissues exhibited increased contractility in the presence of endothelin-1 and larger basal mechanical tone in a unique assay developed to measure the contractile tone of the engineered valve tissues. This finding is important, because it implies that the functional properties of the valve are sensitive to EMT. Our results suggest that cyclic mechanical strain regulates EMT in a strain magnitude and directionally dependent manner.


Asunto(s)
Endotelio/embriología , Válvulas Cardíacas/embriología , Mesodermo/embriología , Morfogénesis , Estrés Mecánico , Actinas/metabolismo , Animales , Anisotropía , Núcleo Celular/metabolismo , Endotelio/metabolismo , Válvulas Cardíacas/metabolismo , Mesodermo/metabolismo , Modelos Biológicos , Contracción Miocárdica , Ovinos , Transducción de Señal , Ingeniería de Tejidos
12.
bioRxiv ; 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38915555

RESUMEN

LMNA -Related Dilated Cardiomyopathy (DCM) is an autosomal-dominant genetic condition with cardiomyocyte and conduction system dysfunction often resulting in heart failure or sudden death. The condition is caused by mutation in the Lamin A/C ( LMNA ) gene encoding Type-A nuclear lamin proteins involved in nuclear integrity, epigenetic regulation of gene expression, and differentiation. Molecular mechanisms of disease are not completely understood, and there are no definitive treatments to reverse progression or prevent mortality. We investigated possible mechanisms of LMNA -Related DCM using induced pluripotent stem cells derived from a family with a heterozygous LMNA c.357-2A>G splice-site mutation. We differentiated one LMNA mutant iPSC line derived from an affected female (Patient) and two non-mutant iPSC lines derived from her unaffected sister (Control) and conducted single-cell RNA sequencing for 12 samples (4 Patient and 8 Control) across seven time points: Day 0, 2, 4, 9, 16, 19, and 30. Our bioinformatics workflow identified 125,554 cells in raw data and 110,521 (88%) high-quality cells in sequentially processed data. Unsupervised clustering, cell annotation, and trajectory inference found complex heterogeneity: ten main cell types; many possible subtypes; and lineage bifurcation for Cardiac Progenitors to Cardiomyocytes (CM) and Epicardium-Derived Cells (EPDC). Data integration and comparative analyses of Patient and Control cells found cell type and lineage differentially expressed genes (DEG) with enrichment to support pathway dysregulation. Top DEG and enriched pathways included: 10 ZNF genes and RNA polymerase II transcription in Pluripotent cells (PP); BMP4 and TGF Beta/BMP signaling, sarcomere gene subsets and cardiogenesis, CDH2 and EMT in CM; LMNA and epigenetic regulation and DDIT4 and mTORC1 signaling in EPDC. Top DEG also included: XIST and other X-linked genes, six imprinted genes: SNRPN , PWAR6 , NDN , PEG10 , MEG3 , MEG8 , and enriched gene sets in metabolism, proliferation, and homeostasis. We confirmed Lamin A/C haploinsufficiency by allelic expression and Western blot. Our complex Patient-derived iPSC model for Lamin A/C haploinsufficiency in PP, CM, and EPDC provided support for dysregulation of genes and pathways, many previously associated with Lamin A/C defects, such as epigenetic gene expression, signaling, and differentiation. Our findings support disruption of epigenomic developmental programs as proposed in other LMNA disease models. We recognized other factors influencing epigenetics and differentiation; thus, our approach needs improvement to further investigate this mechanism in an iPSC-derived model.

13.
Biomed Eng Educ ; : 1-15, 2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-37363619

RESUMEN

In this paper, we altered an in-person high school tissue engineering program to create a virtual course. Through this alteration, we aimed to show that online programs can still be engaging and at the same time provide greater accessibility and flexibility to students. This was achieved through utilizing Google classroom as a virtual platform for students to engage with course modules and assessments. After analyzing pre- and post-program survey responses in both the in-person and online offerings of the CardioStart program, it was found that students improved in their understanding of all of the tissue engineering topics that were introduced in the programs. Furthermore, when comparing the results from the in-person versus online offerings of the program, it was found that the level of student understanding and learning of these topics was similar across the in-person and online programs. We were also able to engage five times the number of students online as compared to the in-person program, which was conducted yearly for six summers. However, many students indicated that their experience would have been better if hands-on activities were included to supplement their knowledge of cell culture techniques after completing the course. The online program improved accessibility and scalability of the program compared to in-person workshops. Future work will consist of bridging this virtual course and the hands-on experiments performed during the in-person program to provide interested students access to laboratory experiences.

14.
Front Physiol ; 14: 1198132, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37601632

RESUMEN

Obstructive sleep apnea (OSA) is a sleep disorder caused by periodic airway obstructions and has been associated with numerous health consequences, which are thought to result from tissue hypoxia. However, challenges in the direct measurement of tissue-level oxygenation make it difficult to analyze the hypoxia exposure pattern in patients. Furthermore, current clinical practice relies on the apnea-hypopnea index (AHI) and pulse oximetry to assess OSA severity, both of which have limitations. To overcome this, we developed a clinically deployable mathematical model, which outputs tissue-level oxygenation. The model incorporates spatial pulmonary oxygen uptake, considers dissolved oxygen, and can use time-dependent patient inputs. It was applied to explore a series of breathing patterns that are clinically differentiated. Supporting previous studies, the result of this analysis indicated that the AHI is an unreliable indicator of hypoxia burden. As a proof of principle, polysomnography data from two patients was analyzed with this model. The model showed greater sensitivity to breathing in comparison with pulse oximetry and provided systemic venous oxygenation, which is absent from clinical measurements. In addition, the dissolved oxygen output was used to calculate hypoxia burden scores for each patient and compared to the clinical assessment, highlighting the importance of event length and cumulative impact of obstructions. Furthermore, an intra-patient statistical analysis was used to underscore the significance of closely occurring obstructive events and to highlight the utility of the model for quantitative data processing. Looking ahead, our model can be used with polysomnography data to predict hypoxic burden on the tissues and help guide patient treatment decisions.

15.
ACS Biomater Sci Eng ; 9(10): 5666-5678, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37713253

RESUMEN

Controlling cell-substrate interactions via the microstructural characteristics of biomaterials offers an advantageous path for modulating cell dynamics, mechanosensing, and migration, as well as for designing immune-modulating implants, all without the drawbacks of chemical-based triggers. Specifically, recent in vivo studies have suggested that a porous implant's microscale curvature landscape can significantly impact cell behavior and ultimately the immune response. To investigate such cell-substrate interactions, we utilized a 3D computational model incorporating the minimum necessary physics of cell migration and cell-substrate interactions needed to replicate known in vitro behaviors. This model specifically incorporates the effect of membrane tension, which was found to be necessary to replicate in vitro cell behavior on curved surfaces. Our simulated substrates represent two classes of porous materials recently used in implant studies, which have markedly different microscale curvature distributions and pore geometries. We found distinct differences between the overall migration behaviors, shapes, and actin polymerization dynamics of cells interacting with the two substrates. These differences were correlated to the shape energy of the cells as they interacted with the porous substrates, in effect interpreting substrate topography as an energetic landscape interrogated by cells. Our results demonstrate that microscale curvature directly influences cell shape and migration and, therefore, is likely to influence cell behavior. This supports further investigation of the relationship between the surface topography of implanted materials and the characteristic immune response, a complete understanding of which would broadly advance principles of biomaterial design.

16.
Front Cell Dev Biol ; 11: 1209542, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37691825

RESUMEN

Altered myofibrillar structure is a consequence of dystrophic pathology that impairs skeletal muscle contractile function and increases susceptibility to contraction injury. In murine Duchenne muscular dystrophy (mdx), myofibrillar alterations are abundant in advanced pathology (>4 months), an age where we formerly established densified microtubule (MT) arrays enriched in detyrosinated (deTyr) tubulin as negative disease modifiers impacting cell mechanics and mechanotransduction. Given the essential role of deTyr-enriched MT arrays in myofibrillar growth, maintenance, and repair, we examined the increased abundance of these arrays as a potential mechanism for these myofibrillar alterations. Here we find an increase in deTyr-tubulin as an early event in dystrophic pathology (4 weeks) with no evidence myofibrillar alterations. At 16 weeks, we show deTyr-enriched MT arrays significantly densified and co-localized to areas of myofibrillar malformation. Profiling the enzyme complexes responsible for deTyr-tubulin, we identify vasohibin 2 (VASH2) and small vasohibin binding protein (SVBP) significantly elevated in the mdx muscle at 4 weeks. Using the genetic increase in VASH2/SVBP expression in 4 weeks wild-type mice we find densified deTyr-enriched MT arrays that co-segregate with myofibrillar malformations similar to those in the 16 weeks mdx. Given that no changes in sarcomere organization were identified in fibers expressing sfGFP as a control, we conclude that disease-dependent densification of deTyr-enriched MT arrays underscores the altered myofibrillar structure in dystrophic skeletal muscle fibers.

17.
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
18.
Biophys Rev (Melville) ; 3(4): 041302, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36407035

RESUMEN

Unbiased evaluation of morphology is crucial to understanding development, mechanics, and pathology of striated muscle tissues. Indeed, the ability of striated muscles to contract and the strength of their contraction is dependent on their tissue-, cellular-, and cytoskeletal-level organization. Accordingly, the study of striated muscles often requires imaging and assessing aspects of their architecture at multiple different spatial scales. While an expert may be able to qualitatively appraise tissues, it is imperative to have robust, repeatable tools to quantify striated myocyte morphology and behavior that can be used to compare across different labs and experiments. There has been a recent effort to define the criteria used by experts to evaluate striated myocyte architecture. In this review, we will describe metrics that have been developed to summarize distinct aspects of striated muscle architecture in multiple different tissues, imaged with various modalities. Additionally, we will provide an overview of metrics and image processing software that needs to be developed. Importantly to any lab working on striated muscle platforms, characterization of striated myocyte morphology using the image processing pipelines discussed in this review can be used to quantitatively evaluate striated muscle tissues and contribute to a robust understanding of the development and mechanics of striated muscles.

19.
Commun Biol ; 5(1): 407, 2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35501466

RESUMEN

Epithelial-mesenchymal Transition (EMT) is a multi-step process that involves cytoskeletal rearrangement. Here, developing and using an image quantification tool, Statistical Parametrization of Cell Cytoskeleton (SPOCC), we have identified an intermediate EMT state with a specific cytoskeletal signature. We have been able to partition EMT into two steps: (1) initial formation of transverse arcs and dorsal stress fibers and (2) their subsequent conversion to ventral stress fibers with a concurrent alignment of fibers. Using the Orientational Order Parameter (OOP) as a figure of merit, we have been able to track EMT progression in live cells as well as characterize and quantify their cytoskeletal response to drugs. SPOCC has improved throughput and is non-destructive, making it a viable candidate for studying a broad range of biological processes. Further, owing to the increased stiffness (and by inference invasiveness) of the intermediate EMT phenotype compared to mesenchymal cells, our work can be instrumental in aiding the search for future treatment strategies that combat metastasis by specifically targeting the fiber alignment process.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias Pulmonares , Citoesqueleto , Transición Epitelial-Mesenquimal/fisiología , Humanos , Neoplasias Pulmonares/genética , Microtúbulos , Fenotipo
20.
Cells ; 10(11)2021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34831422

RESUMEN

The heart has a dynamic mechanical environment contributed by its unique cellular composition and the resultant complex tissue structure. In pathological heart tissue, both the mechanics and cell composition can change and influence each other. As a result, the interplay between the cell phenotype and mechanical stimulation needs to be considered to understand the biophysical cell interactions and organization in healthy and diseased myocardium. In this work, we hypothesized that the overall tissue organization is controlled by varying densities of cardiomyocytes and fibroblasts in the heart. In order to test this hypothesis, we utilized a combination of mechanical strain, co-cultures of different cell types, and inhibitory drugs that block intercellular junction formation. To accomplish this, an image analysis pipeline was developed to automatically measure cell type-specific organization relative to the stretch direction. The results indicated that cardiac cell type-specific densities influence the overall organization of heart tissue such that it is possible to model healthy and fibrotic heart tissue in vitro. This study provides insight into how to mimic the dynamic mechanical environment of the heart in engineered tissue as well as providing valuable information about the process of cardiac remodeling and repair in diseased hearts.


Asunto(s)
Comunicación Celular , Miocardio/patología , Estrés Mecánico , Actinas/metabolismo , Técnicas de Cocultivo , Fibroblastos/patología , Miocitos Cardíacos/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA