Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Ann Biomed Eng ; 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861042

RESUMO

The goal of this work was to develop a general blueprint for a semi-automated image processing tool (SIPT) to measure small, complex features of stent prototypes that can replace the current gold standard of manual measurements. The stents were designed using CAD software and manufactured via laser cutting. Stent prototypes were imaged using a Keyence microscope in top and side view orientations. The SIPT algorithm was developed in MATLAB to extract and measure 4 dimensions of the stent (inner and outer diameter, spring bend outer radius, spring bend width). The same dimensions were also manually measured by an experienced metrology technician as a gold standard comparison. We successfully made over 5000 unique measurements across the 4 key dimensions of 15 stents using the SIPT algorithm. Compared to the gold standard manual method, SIPT reduced measurement time by nearly 90% and increased the total number of measurements captured by over 2300%. The two one-sided test and Bland-Altman analysis demonstrated that SIPT achieved equivalency against the manual method of measurement for all 4 dimensions. In summary, we found that our SIPT software could be used to replace manual measurements and provided substantial time savings with consistent accuracy. Overall, this paper presents a generalizable workflow to isolate and measure critical features of stent prototypes that we believe will provide a valuable, cost-effective tool to other medical device designers seeking to rapidly iterate on unique stent designs or other manufactured parts with small and complex structures.

2.
Biochem Biophys Res Commun ; 686: 149155, 2023 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-37926046

RESUMO

Prosthetic heart valves are associated with almost one quarter of cases of infective endocarditis, a rare but serious condition with a staggering 25 % mortality rate. Without the endothelium of native valves, the risk of infection is exacerbated for implanted devices exposed to blood. There are currently no physiologically relevant in vitro or animal models of prosthetic valve endocarditis (PVE). Of particular importance, Staphylococcus aureus, a common agent of PVE, has demonstrated enhanced binding to blood plasma proteins (e.g., fibrinogen) and exposed matrix under fluid shear stress (FSS). An in vitro platform that mimics the multiple physiological determinants for S. aureus adhesion to prosthetic valve materials would facilitate the discovery of new treatments to minimize PVE. To this end, we developed a first-of-its-kind microphysiological model of PVE to study the effects of several key variables (endothelial cell coverage, fibrinogen deposition, surface treatments, and FSS) on S. aureus adhesion to bioprosthetic material surfaces. Our model demonstrated that viable endothelial monolayers diminished the deposition of fibrinogen and that fibrinogen was required for the subsequent adhesion of S. aureus to the bioprosthetic surface model. Next, we examined factors that affected endothelial cell coverage, such as FSS and glutaraldehyde, a common chemical treatment for bioprosthetic materials. In particular, glutaraldehyde treatment obstructed endothelialization of otherwise biocompatible collagen-coated surfaces, further enabling fibrinogen and S. aureus deposition. In future work, this model could impact multiple research areas, such as screening candidate bioprosthetic valve materials and new surface treatments to prevent PVE and further understanding host-pathogen interactions.


Assuntos
Endocardite Bacteriana , Endocardite , Próteses Valvulares Cardíacas , Animais , Endocardite Bacteriana/microbiologia , Staphylococcus aureus , Próteses Valvulares Cardíacas/efeitos adversos , Aderência Bacteriana , Glutaral , Endocardite/etiologia , Fibrinogênio
3.
Front Mol Biosci ; 10: 1160851, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37577751

RESUMO

Background: Tissue fibrosis is a major healthcare burden that affects various organs in the body for which no effective treatments exist. An underlying, emerging theme across organs and tissue types at early stages of fibrosis is the activation of pericytes and/or fibroblasts in the perivascular space. In hepatic tissue, it is well known that liver sinusoidal endothelial cells (EC) help maintain the quiescence of stellate cells, but whether this phenomenon holds true for other endothelial and perivascular cell types is not well studied. Methods: The goal of this work was to develop an organ-on-chip microvascular model to study the effect of EC co-culture on the activation of perivascular cells perturbed by the pro-fibrotic factor TGFß1. A high-throughput microfluidic platform, PREDICT96, that was capable of imparting physiologically relevant fluid shear stress on the cultured endothelium was utilized. Results: We first studied the activation response of several perivascular cell types and selected a cell source, human dermal fibroblasts, that exhibited medium-level activation in response to TGFß1. We also demonstrated that the PREDICT96 high flow pump triggered changes in select shear-responsive factors in human EC. We then found that the activation response of fibroblasts was significantly blunted in co-culture with EC compared to fibroblast mono-cultures. Subsequent studies with conditioned media demonstrated that EC-secreted factors play at least a partial role in suppressing the activation response. A Luminex panel and single cell RNA-sequencing study provided additional insight into potential EC-derived factors that could influence fibroblast activation. Conclusion: Overall, our findings showed that EC can reduce myofibroblast activation of perivascular cells in response to TGFß1. Further exploration of EC-derived factors as potential therapeutic targets in fibrosis is warranted.

4.
Front Cardiovasc Med ; 9: 993310, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36518682

RESUMO

Introduction: Birth defects, particularly those that affect development of the heart, are a leading cause of morbidity and mortality in infants and young children. Babies born with heart hypoplasia (heart hypoplasia) disorders often have a poor prognosis. It remains unclear whether cardiomyocytes from hypoplastic hearts retain the potential to recover growth, although this knowledge would be beneficial for developing therapies for heart hypoplasia disorders. The objective of this study was to determine the proliferation and maturation potential of cardiomyocytes from hypoplastic hearts and whether these behaviors are influenced by biochemical signaling from the extracellular matrix (ECM) and cyclic mechanical stretch. Method: Congenital diaphragmatic hernia (CDH)-associated heart hypoplasia was induced in rat fetuses by maternal exposure to nitrofen. Hearts were isolated from embryonic day 21 nitrofen-treated fetuses positive for CDH (CDH+) and from fetuses without nitrofen administration during gestation. Results and discussion: CDH+ hearts were smaller and had decreased myocardial proliferation, along with evidence of decreased maturity compared to healthy hearts. In culture, CDH+ cardiomyocytes remained immature and demonstrated increased proliferative capacity compared to their healthy counterparts. Culture on ECM derived from CDH+ hearts led to a significant reduction in proliferation for both CDH+ and healthy cardiomyocytes. Healthy cardiomyocytes were dosed with exogenous nitrofen to examine whether nitrofen may have an aberrant effect on the proliferative ability of cardiomyocyte, yet no significant change in proliferation was observed. When subjected to stretch, CDH+ cardiomyocytes underwent lengthening of sarcomeres while healthy cardiomyocyte sarcomeres were unaffected. Taken together, our results suggest that alterations to environmental cues such as ECM and stretch may be important factors in the pathological progression of heart hypoplasia.

5.
Front Cardiovasc Med ; 9: 884116, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36093160

RESUMO

Characterization of cardiovascular tissue geometry and mechanical properties of large animal models is essential when developing cardiovascular devices such as heart valve replacements. These datasets are especially critical when designing devices for pediatric patient populations, as there is often limited data for guidance. Here, we present a previously unavailable dataset capturing anatomical measurements and mechanical properties of juvenile Yorkshire (YO) and Yucatan (YU) porcine main pulmonary artery (PA) and pulmonary valve (PV) tissue regions that will inform pediatric heart valve design requirements for preclinical animal studies. In addition, we developed a novel radial balloon catheter-based method to measure tissue stiffness and validated it against a traditional uniaxial tensile testing method. YU piglets, which were significantly lower weight than YO counterparts despite similar age, had smaller PA and PV diameters (7.6-9.9 mm vs. 10.1-12.8 mm). Young's modulus (stiffness) was measured for the PA and the PV region using both the radial and uniaxial testing methods. There was no significant difference between the two breeds for Young's modulus measured in the elastic (YU PA 84.7 ± 37.3 kPa, YO PA 79.3 ± 15.7 kPa) and fibrous regimes (YU PA 308.6 ± 59.4 kPa, YO PA 355.7 ± 68.9 kPa) of the stress-strain curves. The two testing techniques also produced similar stiffness measurements for the PA and PV region, although PV data showed greater variation between techniques. Overall, YU and YO piglets had similar PA and PV diameters and tissue stiffness to previously reported infant pediatric patients. These results provide a previously unavailable age-specific juvenile porcine tissue geometry and stiffness dataset critical to the development of pediatric cardiovascular prostheses. Additionally, the data demonstrates the efficacy of a novel balloon catheter-based technique that could be adapted to non-destructively measure tissue stiffness in situ.

6.
Front Cell Infect Microbiol ; 11: 691210, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34123879

RESUMO

Urinary tract infections (UTIs) are among the most common infectious diseases worldwide but are significantly understudied. Uropathogenic E. coli (UPEC) accounts for a significant proportion of UTI, but a large number of other species can infect the urinary tract, each of which will have unique host-pathogen interactions with the bladder environment. Given the substantial economic burden of UTI and its increasing antibiotic resistance, there is an urgent need to better understand UTI pathophysiology - especially its tendency to relapse and recur. Most models developed to date use murine infection; few human-relevant models exist. Of these, the majority of in vitro UTI models have utilized cells in static culture, but UTI needs to be studied in the context of the unique aspects of the bladder's biophysical environment (e.g., tissue architecture, urine, fluid flow, and stretch). In this review, we summarize the complexities of recurrent UTI, critically assess current infection models and discuss potential improvements. More advanced human cell-based in vitro models have the potential to enable a better understanding of the etiology of UTI disease and to provide a complementary platform alongside animals for drug screening and the search for better treatments.


Assuntos
Infecções por Escherichia coli , Infecções Urinárias , Sistema Urinário , Escherichia coli Uropatogênica , Animais , Humanos , Camundongos , Bexiga Urinária
7.
Sci Rep ; 11(1): 12225, 2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34108507

RESUMO

Microphysiological organ-on-chip models offer the potential to improve the prediction of drug safety and efficacy through recapitulation of human physiological responses. The importance of including multiple cell types within tissue models has been well documented. However, the study of cell interactions in vitro can be limited by complexity of the tissue model and throughput of current culture systems. Here, we describe the development of a co-culture microvascular model and relevant assays in a high-throughput thermoplastic organ-on-chip platform, PREDICT96. The system consists of 96 arrayed bilayer microfluidic devices containing retinal microvascular endothelial cells and pericytes cultured on opposing sides of a microporous membrane. Compatibility of the PREDICT96 platform with a variety of quantifiable and scalable assays, including macromolecular permeability, image-based screening, Luminex, and qPCR, is demonstrated. In addition, the bilayer design of the devices allows for channel- or cell type-specific readouts, such as cytokine profiles and gene expression. The microvascular model was responsive to perturbations including barrier disruption, inflammatory stimulation, and fluid shear stress, and our results corroborated the improved robustness of co-culture over endothelial mono-cultures. We anticipate the PREDICT96 platform and adapted assays will be suitable for other complex tissues, including applications to disease models and drug discovery.


Assuntos
Comunicação Celular , Técnicas de Cocultura/métodos , Derme/metabolismo , Endotélio Vascular/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Pericitos/metabolismo , Retina/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas , Derme/citologia , Endotélio Vascular/citologia , Humanos , Pericitos/citologia , Retina/citologia
8.
Crit Rev Biomed Eng ; 44(1-2): 91-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27652453

RESUMO

Between 0.5 and 4 of every 1000 children are born with hydrocephalus. Hydrocephalus is an over-accumulation of cerebrospinal fluid (CSF) in the ventricles of the brain, which can affect cognitive function, vision, appetite, and cranial nerve function. Left untreated, hydrocephalus can result in death. The current treatment for hydrocephalus uses ventriculoperitoneal (VP) shunts with valves to redirect CSF from the ventricles into the peritoneum. Shunt technology is limited by a number of complications, which include infection after implantation, shunt obstruction due to clot formation or catheter obstruction by scar tissue or choroid plexus, disconnection and tubing migration, and overdrainage or underdrainage of CSF due to valve malfunction. While modifications to surgical procedures and shunt design have been introduced, only modest improvements in outcomes have been observed. Here we provide an overview of hydrocephalus, VP shunts, and their modes of failure, and we identify numerous areas of opportunity for biomedical engineers and physicians to collaborate to improve the performance of VP shunts.


Assuntos
Hidrocefalia/cirurgia , Derivação Ventriculoperitoneal , Engenharia Biomédica , Falha de Equipamento , Seguimentos , Humanos , Hidrocefalia/epidemiologia , Melhoria de Qualidade , Falha de Tratamento , Derivação Ventriculoperitoneal/efeitos adversos , Derivação Ventriculoperitoneal/instrumentação , Derivação Ventriculoperitoneal/métodos
9.
Tissue Eng Part A ; 22(3-4): 195-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26573765

RESUMO

Severe birth defects occur in ∼ 2-3% of live-born infants and are a leading cause of death in the young. Structural malformations can occur in just about any major organ system and often their causes are unknown. The pediatric population presents a unique set of opportunities to the field of tissue engineering and regenerative medicine (TERM). Infants and young children have significantly greater regenerative capacity than adults, which could be leveraged in TERM strategies. Children also arguably stand to benefit the most from TERM. Although the lack of growth potential and relatively short life span of synthetic materials may be suitable for adults, it is unacceptable for children. Furthermore, given that there is a particular scarcity of pediatric donor organs, the need for living functional tissue replacements that can grow with the child is quite evident. There is enormous potential for the TERM community to address the needs of the pediatric population.


Assuntos
Anormalidades Múltiplas/terapia , Medicina Regenerativa/métodos , Engenharia Tecidual/métodos , Adolescente , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Medicina Regenerativa/tendências , Engenharia Tecidual/tendências
10.
Adv Healthc Mater ; 4(10): 1545-54, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25988681

RESUMO

Stimulating or maintaining the proliferative capacity of postnatal mammalian cardiomyocytes is a major challenge to cardiac regeneration. Previously, it is found that fetal cardiac extracellular matrix (ECM) can promote neonatal rat cardiomyocyte proliferation in vitro better than neonatal or adult ECM. It is hypothesized that partial digestion of adult ECM (PD-ECM) would liberate less crosslinked components that promote cardiomyocyte proliferation, similar to fetal ECM. Neonatal rat cardiac cells are seeded onto substrates coated with adult rat cardiac ECM that has been solubilized in pepsin-HCl for 1, 3, 6, 12, 24, or 48 h. Cardiomyocyte proliferation and fold-change in numbers from 1 to 5 d are highest on 1 and 3 h PD-ECM compared to other conditions. Sarcomeres tend to mature on 24 and 48 h PD-ECM where low proliferation is observed. 3 h PD-ECM is primarily composed of Fibrillin-1, Fibrinogen, and Laminins while 48 h PD-ECM is dominated by Collagen I. Our results suggest that adult ECM retains regenerative cues that may be masked by more abundant, mature ECM components. PD-ECM provides a simple yet powerful approach to promoting cardiomyocyte proliferation.


Assuntos
Matriz Extracelular/metabolismo , Miócitos Cardíacos/citologia , Sequência de Aminoácidos , Animais , Proliferação de Células , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Feminino , Integrinas/genética , Integrinas/metabolismo , Dados de Sequência Molecular , Miócitos Cardíacos/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Reação em Cadeia da Polimerase , Proteoma/análise , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem
11.
Acta Biomater ; 14: 84-95, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25463503

RESUMO

Solubilized cardiac extracellular matrix (ECM) is being developed as an injectable therapeutic that offers promise for promoting cardiac repair. However, the ECM alone forms a hydrogel that is very soft compared to the native myocardium. As both the stiffness and composition of the ECM are important in regulating cell behavior and can have complex synergistic effects, we sought to develop an ECM-based scaffold with tunable biochemical and mechanical properties. We used solubilized rat cardiac ECM from two developmental stages (neonatal, adult) combined with fibrin hydrogels that were cross-linked with transglutaminase. We show that ECM was retained within the gels and that the Young's modulus could be tuned to span the range of the developing and mature heart. C-kit+ cardiovascular progenitor cells from pediatric patients with congenital heart defects were seeded into the hybrid gels. Both the elastic modulus and composition of the scaffolds impacted the expression of endothelial and smooth muscle cell genes. Furthermore, we demonstrate that the hybrid gels are injectable, and thus have potential for minimally invasive therapies. ECM-fibrin hybrid scaffolds offer new opportunities for exploiting the effects of both composition and mechanical properties in directing cell behavior for tissue engineering.


Assuntos
Matriz Extracelular/metabolismo , Fibrina/farmacologia , Coração/fisiologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Diferenciação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Feminino , Géis/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Injeções , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
12.
Cell Mol Bioeng ; 7(3): 432-445, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25295125

RESUMO

Cardiomyocytes (CMs) undergo a rapid transition from hyperplastic to hypertrophic growth soon after birth, which is a major challenge to the development of engineered cardiac tissue for pediatric patients. Resting membrane potential (Vmem) has been shown to play an important role in cell differentiation and proliferation during development. We hypothesized that depolarization of neonatal CMs would stimulate or maintain CM proliferation in vitro. To test our hypothesis, we isolated postnatal day 3 neonatal rat CMs and subjected them to sustained depolarization via the addition of potassium gluconate or Ouabain to the culture medium. Cell density and CM percentage measurements demonstrated an increase in mitotic CMs along with a ~2 fold increase in CM numbers with depolarization. In addition, depolarization led to an increase in cells in G2 and S phase, indicating increased proliferation, as measured by flow cytometry. Surprisingly depolarization of Vmem with either treatment led to inhibition of proliferation in cardiac fibroblasts. This effect is abrogated when the study was carried out on postnatal day 7 neonatal CMs, which are less proliferative, indicating that the likely mechanism of depolarization is the maintenance of the proliferating CM population. In summary, our findings suggest that depolarization maintains postnatal CM proliferation and may be a novel approach to encourage growth of engineered tissue and cardiac regeneration in pediatric patients.

13.
Biochem Biophys Res Commun ; 439(2): 161-6, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23994333

RESUMO

In this study we present a novel method for studying cellular traction force generation and mechanotransduction in the context of cardiac development. Rat hearts from three distinct stage of development (fetal, neonatal and adult) were isolated, decellularized and characterized via mechanical testing and protein compositional analysis. Stiffness increased ~2-fold between fetal and neonatal time points but not between neonatal and adult. Composition of structural extracellular matrix (ECM) proteins was significantly different between all three developmental ages. ECM that was solubilized via pepsin digestion was cross-linked into polyacrylamide gels of varying stiffness and traction force microscopy was used to assess the ability of mesenchymal stem cells (MSCs) to generate traction stress against the substrates. The response to increasing stiffness was significantly different depending on the developmental age of the ECM. An investigation into early cardiac differentiation of MSCs demonstrated a dependence of the level of expression of early cardiac transcription factors on the composition of the complex ECM. In summary, this study found that complex ECM composition plays an important role in modulating a cell's ability to generate traction stress against a substrate, which is a significant component of mechanotransductive signaling.


Assuntos
Matriz Extracelular/metabolismo , Coração/crescimento & desenvolvimento , Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Animais , Linhagem Celular , Matriz Extracelular/química , Proteínas da Matriz Extracelular/análise , Proteínas da Matriz Extracelular/metabolismo , Mecanotransdução Celular , Células-Tronco Mesenquimais/química , Células-Tronco Mesenquimais/metabolismo , Miocárdio/química , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Estresse Mecânico
14.
Tissue Eng Part A ; 18(9-10): 986-98, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22145703

RESUMO

In pediatric patients requiring vascular reconstruction, the development of a cell-based tissue-engineered vascular patch (TEVP) has great potential to overcome current issues with nonliving graft materials. Determining the optimal cell source is especially critical to TEVP success. In this study, we compared the ability of human aortic smooth muscle cells (HuAoSMCs) and human mesenchymal stem cells (hMSCs) to form cell sheets on thermoresponsive poly(N-isopropylacrylamide) (PIPAAm) substrates. hMSCs treated with transforming growth factor beta 1 (TGFß1) and ascorbic acid (AA) had higher expression of SMC-specific proteins compared to HuAoSMCs. hMSCs also had larger cell area and grew to confluence more quickly on PIPAAm than did HuAoSMCs. hMSCs typically formed cell sheets in 2-3 weeks and had greater wet tissue weight and collagen content compared with HuAoSMC sheets, which generally required growth for up to 5 weeks. Assays for calcification and alkaline phosphatase activity revealed that the osteogenic potential of TGFß1+AA-treated hMSCs was low; however, Alcian Blue staining suggested high chondrogenic behavior of TGFß1+AA-treated hMSCs. Although hMSCs are promising for cell-based TEVPs in their ability to form robust tissue with significant extracellular matrix content, improved control over hMSC behavior will be required for long-term TEVP success.


Assuntos
Células-Tronco Mesenquimais/citologia , Miócitos de Músculo Liso/citologia , Engenharia Tecidual/métodos , Acrilamidas/química , Resinas Acrílicas , Ácido Ascórbico/farmacologia , Western Blotting , Condrogênese/efeitos dos fármacos , Condrogênese/fisiologia , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Polímeros/química , Alicerces Teciduais/química , Fator de Crescimento Transformador beta1/farmacologia
15.
Biomaterials ; 32(24): 5625-32, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21601276

RESUMO

Children suffering from congenital heart defects (CHD) often require vascular reconstruction. Pediatric patients would greatly benefit from a cell-based tissue engineered vascular patch (TEVP) that has potential for growth. As artery structure and function are intimately linked, mimicking native tissue organization is an important design consideration. In this study, we cultured human mesenchymal stem cell on patterned thermo-responsive substrates. Cell alignment improved over time up to 2 wk in culture when sheets were ready for harvest. We then used cell sheets as "functional units" to build complex tissue structures that mimic native vascular smooth muscle cell organization in the medial layer of the artery. Cell sheets could be stacked using a gelatin stamp such that individual sheets in the construct were well aligned with each other (mimic of circumferential orientation) or at angles with respect to each other (mimic of herringbone structure). Controlling tissue organization layer-by-layer will be a powerful approach to building tissues with well defined and complex structure.


Assuntos
Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Acrilamidas , Resinas Acrílicas , Células Cultivadas , Humanos , Microscopia de Contraste de Fase , Polímeros
16.
Biomaterials ; 32(2): 410-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20858564

RESUMO

In the healthy artery, contractile vascular smooth muscle cells (VSMCs) have an elongated shape and are highly aligned but transition to a synthetic phenotype in culture, while additionally becoming well spread and randomly organized. Thus, controlling VSMC phenotype is a challenge in tissue engineering. In this study, we investigated the effects of micropatterning on contractile protein expression in VSMCs at low and high passage and in the presence of transforming growth factor beta 1 (TGFß1). Micropatterning led to significantly decreased cell area, increased elongation, and increased alignment compared to non-patterned VSMCs independent of passage number. In the presence of serum, micropatterning led to increased smooth muscle myosin heavy chain (SM-MHC) and α-actin expression in low passage VSMCs, but had no effect on high passage VSMCs. Micropatterning was as effective as TGFß1 in up-regulating SM-MHC at low passage; however, micropatterning limited VSMC response to TGFß1 at both low and high passage. Investigation of TGFß receptor 1 revealed higher expression in non-patterned VSMCs compared to patterned at high passage. Our studies demonstrate that micropatterning is an important regulator of SM-MHC expression in contractile VSMCs and that it may provide a mechanism for phenotype stabilization in the presence of growth factors.


Assuntos
Forma Celular/efeitos dos fármacos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Western Blotting , Técnicas de Cultura de Células , Masculino , Polímeros/química , Coelhos , Engenharia Tecidual/métodos
17.
J Cell Physiol ; 225(1): 115-22, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20648629

RESUMO

Vascular disease, such as atherosclerosis, is accompanied by changes in the mechanical properties of the vessel wall. Although altered mechanics is thought to contribute to disease progression, the molecular mechanisms whereby vessel wall stiffening could promote vascular occlusive disease remain unclear. It is well known that platelet-derived growth factor (PDGF) is a major stimulus for the abnormal migration and proliferation of vascular smooth muscle cells (VSMCs) and contributes critically to vascular disease. Here we used engineered substrates with tunable mechanical properties to explore the effect of tissue stiffness on PDGF signaling in VSMCs as a potential mechanism whereby vessel wall stiffening could promote vascular disease. We found that substrate stiffness significantly enhanced PDGFR activity and VSMC proliferation. After ligand binding, PDGFR followed distinct routes of activation in cells cultured on stiff versus soft substrates, as demonstrated by differences in its intensity and duration of activation, sensitivity to cholesterol extracting agent, and plasma membrane localization. Our results suggest that stiffening of the vessel wall could actively promote pathogenesis of vascular disease by enhancing PDGFR signaling to drive VSMC growth and survival.


Assuntos
Aterosclerose/metabolismo , Técnicas de Cultura de Células/instrumentação , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Resinas Acrílicas/química , Animais , Aterosclerose/patologia , Bovinos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Elasticidade , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo
18.
Biomaterials ; 29(17): 2565-72, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18377979

RESUMO

The proper function of many tissues depends critically on the structural organization of the cells and matrix of which they are comprised. Therefore, in order to engineer functional tissue equivalents that closely mimic the unique properties of native tissues it is necessary to develop strategies for reproducing the complex, highly organized structure of these tissues. To this end, we sought to develop a simple method for generating cell sheets that have defined ECM/cell organization using microtextured, thermoresponsive polystyrene substrates to guide cell organization and tissue growth. The patterns consisted of large arrays of alternating grooves and ridges (50 microm wide, 5 microm deep). Vascular smooth muscle cells cultured on these substrates produced intact sheets consisting of cells that exhibited strong alignment in the direction of the micropattern. These sheets could be readily transferred from patterned substrates to non-patterned substrates without the loss of tissue organization. Ultimately, such sheets will be layered to form larger tissues with defined ECM/cell organization that spans multiple length scales.


Assuntos
Acrilamidas/química , Materiais Biocompatíveis/química , Biotecnologia/métodos , Técnicas de Cultura de Células/métodos , Polímeros/química , Engenharia Tecidual/métodos , Resinas Acrílicas , Adulto , Aorta/citologia , Adesão Celular , Células Cultivadas , Matriz Extracelular/química , Humanos , Microscopia Eletrônica de Varredura , Músculo Liso Vascular/citologia , Propriedades de Superfície , Temperatura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA