Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38606850

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is characterized by a dense fibrotic stroma that contributes to aggressive tumor biology and therapeutic resistance. Current in vitro PDAC models lack sufficient optical and physical access for fibrous network visualization, in situ mechanical stiffness measurement, and metabolomic profiling. Here, we describe an openable multilayer microfluidic PDAC-on-a-chip platform that consists of pancreatic tumor cells (PTCs) and pancreatic stellate cells (PSCs) embedded in a 3D collagen matrix that mimics the stroma. Our system allows fibrous network visualization via reflected light confocal (RLC) microscopy, in situ mechanical stiffness testing using atomic force microscopy (AFM), and compartmentalized hydrogel extraction for PSC metabolomic profiling via mass spectrometry (MS) analysis. In comparing cocultures of gel-embedded PSCs and PTCs with PSC-only monocultures, RLC microscopy identified a significant decrease in pore size and corresponding increase in fiber density. In situ AFM indicated significant increases in stiffness, and hallmark characteristics of PSC activation were observed using fluorescence microscopy. PSCs in coculture also demonstrated localized fiber alignment and densification as well as increased collagen production. Finally, an untargeted MS study putatively identified metabolic contributions consistent with in vivo PDAC studies. Taken together, this platform can potentially advance our understanding of tumor-stromal interactions toward the discovery of novel therapies.

2.
Cell Rep Med ; 5(5): 101520, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38642550

RESUMO

Pathogenic variants in MYH7 and MYBPC3 account for the majority of hypertrophic cardiomyopathy (HCM). Targeted drugs like myosin ATPase inhibitors have not been evaluated in children. We generate patient and variant-corrected iPSC-cardiomyocytes (CMs) from pediatric HCM patients harboring single variants in MYH7 (V606M; R453C), MYBPC3 (G148R) or digenic variants (MYBPC3 P955fs, TNNI3 A157V). We also generate CMs harboring MYBPC3 mono- and biallelic variants using CRISPR editing of a healthy control. Compared with isogenic and healthy controls, variant-positive CMs show sarcomere disorganization, higher contractility, calcium transients, and ATPase activity. However, only MYH7 and biallelic MYBPC3 variant-positive CMs show stronger myosin-actin binding. Targeted myosin ATPase inhibitors show complete rescue of the phenotype in variant-positive CMs and in cardiac Biowires to mirror isogenic controls. The response is superior to verapamil or metoprolol. Myosin inhibitors can be effective in genotypically diverse HCM highlighting the need for myosin inhibitor drug trials in pediatric HCM.


Assuntos
Miosinas Cardíacas , Cardiomiopatia Hipertrófica , Células-Tronco Pluripotentes Induzidas , Miócitos Cardíacos , Cadeias Pesadas de Miosina , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/tratamento farmacológico , Cardiomiopatia Hipertrófica/patologia , Cardiomiopatia Hipertrófica/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Criança , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Genótipo , Miosinas/metabolismo , Miosinas/genética , Masculino , Feminino , Sarcômeros/metabolismo , Sarcômeros/genética
3.
Stem Cell Res Ther ; 14(1): 96, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37076906

RESUMO

BACKGROUND: Constructs currently used to repair or replace congenitally diseased pediatric heart valves lack a viable cell population capable of functional adaptation in situ, necessitating repeated surgical intervention. Heart valve tissue engineering (HVTE) can address these limitations by producing functional living tissue in vitro that holds the potential for somatic growth and remodelling upon implantation. However, clinical translation of HVTE strategies requires an appropriate source of autologous cells that can be non-invasively harvested from mesenchymal stem cell (MSC)-rich tissues and cultured under serum- and xeno-free conditions. To this end, we evaluated human umbilical cord perivascular cells (hUCPVCs) as a promising cell source for in vitro production of engineered heart valve tissue. METHODS: The proliferative, clonogenic, multilineage differentiation, and extracellular matrix (ECM) synthesis capacities of hUCPVCs were evaluated in a commercial serum- and xeno-free culture medium (StemMACS™) on tissue culture polystyrene and benchmarked to adult bone marrow-derived MSCs (BMMSCs). Additionally, the ECM synthesis potential of hUCPVCs was evaluated when cultured on polycarbonate polyurethane anisotropic electrospun scaffolds, a representative biomaterial for in vitro HVTE. RESULTS: hUCPVCs had greater proliferative and clonogenic potential than BMMSCs in StemMACS™ (p < 0.05), without differentiation to osteogenic and adipogenic phenotypes associated with valve pathology. Furthermore, hUCPVCs cultured with StemMACS™ on tissue culture plastic for 14 days synthesized significantly more total collagen, elastin, and sulphated glycosaminoglycans (p < 0.05), the ECM constituents of the native valve, than BMMSCs. Finally, hUCPVCs retained their ECM synthesizing capacity after 14 and 21 days in culture on anisotropic electrospun scaffolds. CONCLUSION: Overall, our findings establish an in vitro culture platform that uses hUCPVCs as a readily-available and non-invasively sourced autologous cell population and a commercial serum- and xeno-free culture medium to increase the translational potential of future pediatric HVTE strategies. This study evaluated the proliferative, differentiation and extracellular matrix (ECM) synthesis capacities of human umbilical cord perivascular cells (hUCPVCs) when cultured in serum- and xeno-free media (SFM) against conventionally used bone marrow-derived MSCs (BMMSCs) and serum-containing media (SCM). Our findings support the use of hUCPVCs and SFM for in vitro heart valve tissue engineering (HVTE) of autologous pediatric valve tissue. Figure created with BioRender.com.


Assuntos
Células-Tronco Mesenquimais , Engenharia Tecidual , Adulto , Humanos , Criança , Cordão Umbilical , Diferenciação Celular , Meios de Cultura , Células Cultivadas , Proliferação de Células
4.
Nat Commun ; 13(1): 6166, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36257954

RESUMO

The intercalated disc (ICD) is a unique membrane structure that is indispensable to normal heart function, yet its structural organization is not completely understood. Previously, we showed that the ICD-bound transmembrane protein 65 (Tmem65) was required for connexin43 (Cx43) localization and function in cultured mouse neonatal cardiomyocytes. Here, we investigate the functional and cellular effects of Tmem65 reductions on the myocardium in a mouse model by injecting CD1 mouse pups (3-7 days after birth) with recombinant adeno-associated virus 9 (rAAV9) harboring Tmem65 shRNA, which reduces Tmem65 expression by 90% in mouse ventricles compared to scrambled shRNA injection. Tmem65 knockdown (KD) results in increased mortality which is accompanied by eccentric hypertrophic cardiomyopathy within 3 weeks of injection and progression to dilated cardiomyopathy with severe cardiac fibrosis by 7 weeks post-injection. Tmem65 KD hearts display depressed hemodynamics as measured echocardiographically as well as slowed conduction in optical recording accompanied by prolonged PR intervals and QRS duration in electrocardiograms. Immunoprecipitation and super-resolution microscopy demonstrate a physical interaction between Tmem65 and sodium channel ß subunit (ß1) in mouse hearts and this interaction appears to be required for both the establishment of perinexal nanodomain structure and the localization of both voltage-gated sodium channel 1.5 (NaV1.5) and Cx43 to ICDs. Despite the loss of NaV1.5 at ICDs, whole-cell patch clamp electrophysiology did not reveal reductions in Na+ currents but did show reduced Ca2+ and K+ currents in Tmem65 KD cardiomyocytes in comparison to control cells. We conclude that disrupting Tmem65 function results in impaired ICD structure, abnormal cardiac electrophysiology, and ultimately cardiomyopathy.


Assuntos
Conexina 43 , Canal de Sódio Disparado por Voltagem NAV1.5 , Camundongos , Animais , Conexina 43/genética , Conexina 43/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , RNA Interferente Pequeno/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/metabolismo
5.
Acta Biomater ; 147: 209-220, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35643199

RESUMO

Common periodontal disease treatment procedures often fail to restore the structural integrity of the junctional epithelium (JE), the epithelial attachment of the gum to the tooth, leaving the tooth-gum interface prone to bacterial colonization. To address this issue, we introduced a novel bio-inspired protein complex comprised of a proline-rich enamel protein, SCPPPQ1, and laminin 332 (LAM332) to enhance the JE attachment. Using quartz crystal microbalance with dissipation monitoring (QCM-D), we showed that SCPPPQ1 and LAM332 interacted and assembled into a protein complex with high-affinity adsorption of 5.9e-8 [M] for hydroxyapatite (HA), the main component of the mineralized tooth surfaces. We then designed a unique shear device to study the adhesion strength of the oral epithelial cells to HA. The SCPPPQ1/LAM332 complex resulted in a twofold enhancement in adhesion strength of the cells to HA compared to LAM332 (from 31 dyn/cm2 to 63 dyn/cm2). In addition, using a modified wound-healing assay, we showed that gingival epithelial cells demonstrated a significantly high migration rate of 2.7 ± 0.24 µm/min over SCPPPQ1/LAM332-coated surfaces. Our collective data show that this protein complex has the potential to be further developed in designing a bioadhesive to enhance the JE attachment and protect the underlying connective tissue from bacterial invasion. However, its efficacy for wound healing requires further testing in vivo. STATEMENT OF SIGNIFICANCE: This work is the first functional study towards understanding the combined role of the enamel protein SCPPPQ1 and laminin 332 (LAM332) in the epithelial attachment of the gum, the junctional epithelium (JE), to the tooth hydroxyapatite surfaces. Such studies are essential for developing therapeutic approaches to restore the integrity of the JE in the destructive form of gum infection. We have developed a model system that provided the first evidence of the strong interaction between SCPPPQ1 and LAM332 on hydroxyapatite surfaces that favored protein adsorption and subsequently oral epithelial cell attachment and migration. Our collective data strongly suggested using the SCPPPQ1/LAM332 complex to accelerate the reestablishment of the JE after surgical gum removal to facilitate gum regeneration.


Assuntos
Inserção Epitelial , Células Epiteliais , Membrana Basal/metabolismo , Inserção Epitelial/metabolismo , Gengiva , Hidroxiapatitas , Regeneração , Cicatrização
6.
Ann Biomed Eng ; 50(9): 1073-1089, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35622208

RESUMO

Repair and replacement solutions for congenitally diseased heart valves capable of post-surgery growth and adaptation have remained elusive. Tissue engineered heart valves (TEHVs) offer a potential biological solution that addresses the drawbacks of existing valve replacements. Typically, TEHVs are made from thin, fibrous biomaterials that either become cell populated in vitro or in situ. Often, TEHV designs poorly mimic the anisotropic mechanical properties of healthy native valves leading to inadequate biomechanical function. Mechanical conditioning of engineered tissues with anisotropic strain application can induce extracellular matrix remodelling to alter the anisotropic mechanical properties of a construct, but implementation has been limited to small-scale set-ups. To address this limitation for TEHV applications, we designed and built a mechanobioreactor capable of modulating biaxial strain anisotropy applied to large, thin, biomaterial sheets in vitro. The bioreactor can independently control two orthogonal stretch axes to modulate applied strain anisotropy on biomaterial sheets from 13 × 13 mm2 to 70 × 40 mm2. A design of experiments was performed using experimentally validated finite element (FE) models and demonstrated that biaxial strain was applied uniformly over a larger percentage of the cell seeded area for larger sheets (13 × 13 mm2: 58% of sheet area vs. 52 × 31 mm2: 86% of sheet area). Furthermore, bioreactor prototypes demonstrated that over 70% of the cell seeding area remained uniformly strained under different prescribed protocols: equibiaxial amplitudes between 5 to 40%, cyclic frequencies between 0.1 to 2.5 Hz and anisotropic strain ratios between 0:1 (constrained uniaxial) to 2:1. Lastly, proof-of-concept experiments were conducted where we applied equibiaxial (εx = εy = 8.75%) and anisotropic (εx = 12.5%, εy = 5%) strain protocols to cell-seeded, electrospun scaffolds. Cell nuclei and F-actin aligned to the vector-sum strain direction of each prescribed protocol (nuclei alignment: equibiaxial: 43.2° ± 1.8°, anisotropic: 17.5° ± 1.7°; p < 0.001). The abilities of this bioreactor to prescribe different strain amplitude, frequency and strain anisotropy protocols to cell-seeded scaffolds will enable future studies into the effects of anisotropic loading protocols on mechanically conditioned TEHVs and other engineered planar connective tissues.


Assuntos
Materiais Biocompatíveis , Engenharia Tecidual , Anisotropia , Matriz Extracelular , Valvas Cardíacas , Estresse Mecânico , Engenharia Tecidual/métodos
7.
Circulation ; 145(18): 1412-1426, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35089805

RESUMO

BACKGROUND: Human pluripotent stem cell (hPSC)-derived cardiomyocytes (hPSC-CMs) have tremendous promise for application in cardiac regeneration, but their translational potential is limited by an immature phenotype. We hypothesized that large-scale manufacturing of mature hPSC-CMs could be achieved through culture on polydimethylsiloxane (PDMS)-lined roller bottles and that the transplantation of these cells would mediate better structural and functional outcomes than with conventional immature hPSC-CM populations. METHODS: We comprehensively phenotyped hPSC-CMs after in vitro maturation for 20 and 40 days on either PDMS or standard tissue culture plastic substrates. All hPSC-CMs were generated from a transgenic hPSC line that stably expressed a voltage-sensitive fluorescent reporter to facilitate in vitro and in vivo electrophysiological studies, and cardiomyocyte populations were also analyzed in vitro by immunocytochemistry, ultrastructure and fluorescent calcium imaging, and bulk and single-cell transcriptomics. We next compared outcomes after the transplantation of these populations into a guinea pig model of myocardial infarction using end points including histology, optical mapping of graft- and host-derived action potentials, echocardiography, and telemetric electrocardiographic monitoring. RESULTS: We demonstrated the economic generation of >1×108 mature hPSC-CMs per PDMS-lined roller bottle. Compared with their counterparts generated on tissue culture plastic substrates, PDMS-matured hPSC-CMs exhibited increased cardiac gene expression and more mature structural and functional properties in vitro. More important, intracardiac grafts formed with PDMS-matured myocytes showed greatly enhanced structure and alignment, better host-graft electromechanical integration, less proarrhythmic behavior, and greater beneficial effects on contractile function. CONCLUSIONS: We describe practical methods for the scaled generation of mature hPSC-CMs and provide the first evidence that the transplantation of more mature cardiomyocytes yields better outcomes in vivo.


Assuntos
Miócitos Cardíacos , Células-Tronco Pluripotentes , Animais , Diferenciação Celular , Linhagem Celular , Cobaias , Humanos , Miócitos Cardíacos/metabolismo , Plásticos/metabolismo , Células-Tronco Pluripotentes/metabolismo
8.
Eur J Cardiothorac Surg ; 61(2): 367-375, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-34718497

RESUMO

OBJECTIVES: The effect of aortic haemodynamics on arterial wall properties in ascending thoracic aortic aneurysms (ATAAs) is not well understood. We aim to delineate the relationship between shear forces along the aortic wall and loco-regional biomechanical properties associated with the risk of aortic dissection. METHODS: Five patients with ATAA underwent preoperative magnetic resonance angiogram and four-dimensional magnetic resonance imaging. From these scans, haemodynamic models were constructed to estimate maximum wall shear stress (WSS), maximum time-averaged WSS, average oscillating shear index and average relative residence time. Fourteen resected aortic samples from these patients underwent bi-axial tensile testing to determine energy loss (ΔUL) and elastic modulus (E10) in the longitudinal (ΔULlong, E10long) and circumferential (ΔULcirc, E10circ) directions and the anisotropic index (AI) for each parameter. Nine resected aortic samples underwent peel testing to determine the delamination strength (Sd). Haemodynamic indices were then correlated to the biomechanical properties. RESULTS: A positive correlation was found between maximum WSS and ΔULlong rs=0.75, P = 0.002 and AIΔUL (rs=0.68, P=0.01). Increasing maximum time-averaged WSS was found to be associated with increasing ΔULlong (rs=0.73, P = 0.003) and AIΔUL (rs=0.62, P=0.02). Average oscillating shear index positively correlated with Sd (rs=0.73,P=0.04). No significant relationship was found between any haemodynamic index and E10, or between relative residence time and any biomechanical property. CONCLUSIONS: Shear forces at the wall of ATAAs are associated with local degradation of arterial wall viscoelastic hysteresis (ΔUL) and delamination strength, a surrogate for aortic dissection. Haemodynamic indices may provide insights into aortic wall integrity, ultimately leading to novel metrics for assessing risks associated with ATAAs.


Assuntos
Aneurisma da Aorta Torácica , Aneurisma Aórtico , Dissecção Aórtica , Aorta , Fenômenos Biomecânicos , Hemodinâmica , Humanos , Estresse Mecânico
10.
J Mech Behav Biomed Mater ; 115: 104245, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33310684

RESUMO

Cells sense and respond to the heterogeneous mechanical properties of their tissue microenvironment, with implications for the development of many diseases, including cancer, fibrosis, and aortic valve disease. Characterization of tissue mechanical heterogeneity on cellular length scales of tens of micrometers is thus important for understanding disease mechanobiology. In this study, we developed a low-cost bench-top microindentation system to readily map focal microscale soft tissue mechanical properties. The device was validated by comparison with atomic force microscopy nanoindentation of polyacrylamide gels. To demonstrate its utility, the device was used to measure the focal microscale elastic moduli of normal and diseased porcine aortic valve leaflet tissue. Consistent with previous studies, the fibrosa layer of intact leaflets was found to be 1.91-fold stiffer than the ventricularis layer, with both layers exhibiting significant heterogeneity in focal elastic moduli. For the first time, the microscale compressive moduli of focal proteoglycan-rich lesions in the fibrosa of early diseased porcine aortic valve leaflets were measured and found to be 2.44-fold softer than those of normal tissue. These data provide new insights into the tissue micromechanical environment in valvular disease and demonstrate the utility of the microindentation device for facile measurement of the focal mechanical properties of soft tissues.


Assuntos
Valva Aórtica , Animais , Fenômenos Biomecânicos , Microscopia de Força Atômica , Pressão , Estresse Mecânico , Suínos
11.
Biomed Mater ; 16(3)2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33086195

RESUMO

One aspect of the challenge of engineering viable tissuesex vivois the generation of perfusable microvessels of varying diameters. In this work, we take the approach of using hydrogel-based microfluidics seeded with endothelial cells (ECs) to form small artery/vein-like vessels, in conjunction with using the self-assembly behavior of ECs to form capillary-like vessels when co-cultured with multipotent stromal cells (MSCs). In exploring this approach, we focused on investigating collagen, fibrin, and various collagen-fibrin co-gel formulations for their potential suitability as serving as scaffold materials by surveying their angiogencity and mechanical properties. Fibrin and co-gels successfully facilitated multicellular EC sprouting, whereas collagen elicited a migration response of individual ECs, unless supplemented with the protein kinase C (PKC)-activator, phorbol 12-myristate 13-acetate. Collagen scaffolds were also found to severely contract when embedded with mesenchymal cells, but this contraction could be abrogated with the addition of fibrin. Increasing collagen content within co-gel formulations, however, imparted a higher compressive modulus and allowed for the reliable formation of intact hydrogel-based microchannels which could then be perfused. Given the bioactivity and mechanical benefits of fibrin and collagen, respectively, collagen-fibrin co-gels are a promising scaffold option for generating vascularized tissue constructs.


Assuntos
Fibrina , Células-Tronco Mesenquimais , Colágeno/metabolismo , Células Endoteliais/fisiologia , Hidrogéis , Células-Tronco Mesenquimais/metabolismo , Microfluídica , Morfogênese , Neovascularização Fisiológica , Engenharia Tecidual/métodos
12.
J Am Heart Assoc ; 9(15): e016715, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32750292

RESUMO

Background Current methods for aortic dissection risk assessment are inadequate for patients with ascending aortic aneurysms associated with either bicuspid aortic valves (BAVs) or tricuspid aortic valves (TAVs). Biomechanical testing of aortic tissue may provide novel insights and biomarkers. Methods and Results From March 2017 to August 2019, aneurysmal ascending aortas (BAV=23, TAV=23) were collected from elective aortic surgery, normal aortas from transplant donors (n=9), and dissected aortas from surgery for aortic dissection (n=7). These aortas underwent delamination testing in simulation of aortic dissection. Biaxial tensile testing was performed to determine modulus of elasticity (aortic stiffness), and energy loss (a measure of efficiency in performing the Windkessel function). Delamination strength (Sd) was lowest in dissected aortas (18±6 mN/mm) and highest in normal aortas (58±16 mN/mm), and aneurysms fell in between, with greater Sd in the BAV group (37±10 mN/mm) than the TAV group (27±10 mN/mm) (P<0.001). Bicuspid aortopathy was associated with greater stiffness (P<0.001), while aneurysms with TAV demonstrated greater energy loss (P<0.001). Sd decreased by 7.8±1.2 mmol/L per mm per decade of life (r2=0.45, P<0.001), and it was significantly lower for patients with hypertension (P=0.001). Sd decreased by 6.1±2.1 mmol/L per mm with each centimeter increase in aortic diameter (r2=0.15, P=0.007). Increased energy loss was associated with decreased Sd (r2=0.41), whereas there was no relationship between Sd and aortic stiffness. Conclusions Aneurysms with BAV had higher Sd than those with TAV, suggesting that BAV was protective. Energy loss was lower in aneurysms with BAV, and inversely associated with Sd, representing a potential novel biomarker.


Assuntos
Dissecção Aórtica/etiologia , Doença da Válvula Aórtica Bicúspide/complicações , Adulto , Idoso , Dissecção Aórtica/patologia , Aorta/patologia , Doença da Válvula Aórtica Bicúspide/fisiopatologia , Fenômenos Biomecânicos , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
13.
Arterioscler Thromb Vasc Biol ; 40(7): 1763-1776, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32493168

RESUMO

OBJECTIVE: Vascular calcification is a pathology characterized by arterial mineralization, which is a common late-term complication of atherosclerosis that independently increases the risk of adverse cardiovascular events by fourfold. A major source of calcifying cells is transdifferentiating vascular smooth muscle cells (VSMCs). Previous studies showed that deletion of the collagen-binding receptor, DDR1 (discoidin domain receptor-1), attenuated VSMC calcification. Increased matrix stiffness drives osteogenesis, and DDR1 has been implicated in stiffness sensing in other cell types; however, the role of DDR1 as a mechanosensor in VSMCs has not been investigated. Here, we test the hypothesis that DDR1 senses increased matrix stiffness and promotes VSMC transdifferentiation and calcification. Approach and Results: Primary VSMCs isolated from Ddr1+/+ (wild-type) and Ddr1-/- (knockout) mice were studied on collagen-I-coated silicon substrates of varying stiffness, culturing in normal or calcifying medium. DDR1 expression and phosphorylation increased with increasing stiffness, as did in vitro calcification, nuclear localization of Runx2 (Runt-related transcription factor 2), and expression of other osteochondrocytic markers. By contrast, DDR1 deficient VSMCs were not responsive to stiffness and did not undergo transdifferentiation. DDR1 regulated stress fiber formation and RhoA (ras homolog family member A) activation through the RhoGEF (rho guanine nucleotide exchange factor), Vav2. Inhibition of actomyosin contractility reduced Runx2 activation and attenuated in vitro calcification in wild-type VSMCs. Finally, a novel positive feedforward loop was uncovered between DDR1 and actomyosin contractility, important in regulating DDR1 expression, clustering, and activation. CONCLUSIONS: This study provides mechanistic insights into DDR1 mechanosignaling and shows that DDR1 activity and actomyosin contractility are interdependent in mediating stiffness-dependent increases in VSMC calcification.


Assuntos
Aterosclerose/enzimologia , Transdiferenciação Celular , Receptor com Domínio Discoidina 1/metabolismo , Matriz Extracelular/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Osteogênese , Calcificação Vascular/enzimologia , Proteína rhoA de Ligação ao GTP/metabolismo , Actomiosina/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Receptor com Domínio Discoidina 1/deficiência , Receptor com Domínio Discoidina 1/genética , Modelos Animais de Doenças , Matriz Extracelular/patologia , Mecanotransdução Celular , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/patologia
14.
Acta Biomater ; 103: 129-141, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31821896

RESUMO

In vascular tissue engineering, the ability to obtain a robust and safe vascular tissue cell source (e.g. vascular smooth muscle cells (VSMCs)) and to promote vascular tissue-specific extracellular matrix (ECM) protein production is critically important. Mature blood vessel-derived VSMCs are not practical for in vitro vascular tissue regeneration. The authors have conceived a strategy to differentiate adipose derived stromal cells (ASCs) into VSMC-like cells (ASC-VSMCs) that were similar to mature umbilical artery VSMCs at the transcriptional, protein and contraction function levels. Monocytes/macrophages are known as important regulators of the inflammation and regeneration processes within different tissue types of the body. However, our understanding of the potential interactions between specific tissue-like cells differentiated from stem/stromal cells (e.g. ASC-VSMCs) and monocytes/macrophages (cued by specific biomaterial scaffolds) is still limited. In this study, indirect and direct ASC-VSMC-monocyte co-cultures were constructed within a porous polyurethane scaffold (D-PHI) previously shown to have an immunomodulatory character. The effects of monocytes/macrophages on the cellularity (cell number detected with DNA quantification assay), ECM (glycosaminoglycan (GAG), collagen, and elastin) accumulation as well as the maintenance of contractile VSMC markers (calponin and smoothelin) of the ASC-VSMCs after a month of co-culture were investigated. It was found that monocyte paracrine signalling in D-PHI positively affected the cellularity and ECM accumulation of ASC-VSMCs in co-culture. Cause-effect relationships were also identified between the release of pro-inflammatory/anti-inflammatory factors (i.e. IL6, TGF-ß1) in co-culture and the expression of contractile proteins (calponin and smoothelin) by ASC-VSMCs. This study demonstrated the importance of combining an immune cell strategy with stromal cell derived VSMCs (i.e. ASC-VSMCs) to achieve a practical vascular tissue engineering outcome. STATEMENT OF SIGNIFICANCE: Adipose stromal cell derived-vascular smooth muscle cells (ASC-VSMCs) are a promising cell source for vascular tissue engineering. Monocytes/monocyte derived macrophages can be harnessed as an immune-assisted strategy to promote vascular tissue regeneration. This study demonstrated that the co-culture of human ASC-VSMCs with monocytes significantly enhanced the cellularity and extracellular matrix (ECM) accumulation within anionic polyurethane (D-PHI) scaffolds, partially mediated by monocyte paracrine signalling mechanisms. In addition, specific VSMC contractile markers (calponin and smoothelin) were still present in ASC-VSMCs when the cells were exposed to monocytes for a month in vitro. This study corroborated the potential selection of ASC-VSMCs for in vitro engineering of vascular tissue in an immunomodulatory biomaterial scaffold (e.g. D-PHI) based co-culture system containing monocytes.


Assuntos
Tecido Adiposo/citologia , Matriz Extracelular/metabolismo , Monócitos/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Comunicação Parácrina , Transdução de Sinais , Biomarcadores/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Citocinas/metabolismo , Proteínas do Citoesqueleto/metabolismo , DNA/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Interleucina-6/metabolismo , Proteínas dos Microfilamentos/metabolismo , Contração Muscular , Proteínas Musculares/metabolismo , Fenótipo , Células Estromais , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Calponinas
15.
Biotechnol J ; 15(3): e1900118, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31657515

RESUMO

While extracellular matrix (ECM)-derived coatings have the potential to direct the response of cell populations in culture, there is a need to investigate the effects of ECM sourcing and processing on substrate bioactivity. To develop improved cell culture models for studying adipogenesis, the current study examines the proliferation and adipogenic differentiation of human adipose-derived stem/stromal cells (ASCs) on a range of ECM-derived coatings. Human decellularized adipose tissue (DAT) and commercially available bovine tendon collagen (COL) are digested with α-amylase or pepsin to prepare the coatings. Physical characterization demonstrates that α-amylase digestion generates softer, thicker, and more stable coatings, with a fibrous tissue-like ultrastructure that is lost in the pepsin-digested thin films. ASCs cultured on the α-amylase-digested ECM have a more spindle-shaped morphology, and proliferation is significantly enhanced on the α-amylase-digested DAT coatings. Further, the α-amylase-digested DAT provides a more pro-adipogenic microenvironment, based on higher levels of adipogenic gene expression, glycerol-3-phosphate dehydrogenase (GPDH) enzyme activity, and perilipin staining. Overall, this study supports α-amylase digestion as a new approach for generating bioactive ECM-derived coatings, and demonstrates tissue-specific bioactivity using adipose-derived ECM to enhance ASC proliferation and adipogenic differentiation.


Assuntos
Tecido Adiposo/citologia , Tecido Adiposo/enzimologia , alfa-Amilases/metabolismo , Adipogenia/genética , Adipogenia/fisiologia , Tecido Adiposo/ultraestrutura , Animais , Bovinos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Células Cultivadas , Colágeno/química , Glicerol-3-Fosfato Desidrogenase (NAD+)/metabolismo , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Microscopia de Força Atômica , Microscopia Eletroquímica de Varredura , Tendões/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química
16.
Hypertension ; 73(3): 561-570, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30636551

RESUMO

The proto-oncogene c-myb (and corresponding nuclear transcription factor, c-Myb) regulates the proliferation and differentiation of hematologic and vascular smooth muscle cells; however, the role of c-Myb in blood pressure regulation is unknown. Here, we show that mice homozygous for a hypomorphic c-myb allele ( c-myb h/h) conferring reduced c-Myb activity manifest reduced peripheral blood and kidney B220+ B-cells and have decreased systolic (104±2 versus 120±1 mm Hg; P<0.0001) and diastolic blood pressure (71±2 versus 83±1 mm Hg; P<0.0001) compared with WT (wild type) mice. Additionally, c-myb h/h mice had lower susceptibility to deoxycorticosterone acetate-salt experimental hypertension. Although cardiac (echocardiography) and resistance artery (perfusion myography) functions were normal, metabolic cage studies revealed that c-myb h/h mice had increased 24-hour urine output and sodium excretion versus WT. Reconstitution of WT mice with c-myb h/h bone marrow transplant and chimeric bone marrow transplant using mice lacking B-cells ( J H T; h/h>WT and h/h:J H T>WT, respectively) decreased blood pressure and increased 24-hour urine output compared with controls ( WT>WT; WT:J H T>WT). J H T mice also had decreased systolic (103±2 versus 115±1 mm Hg; P<0.0001) and diastolic blood pressure (71±2 versus 79±1; P<0.01) and increased 24-hour urine output versus WT. Real-time quantitative reverse transcription polymerase chain reaction of kidney medulla revealed reduced V2R (vasopressin receptor 2) expression in c-myb h/h and J H T mice. These data implicate B-cells in the regulation of V2R and its associated effects on salt and water handling and blood pressure homeostasis.


Assuntos
Linfócitos B/metabolismo , Pressão Sanguínea/fisiologia , Hipertensão/imunologia , Miócitos de Músculo Liso/metabolismo , Animais , Linfócitos B/patologia , Diferenciação Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/patologia , Proteínas Proto-Oncogênicas c-myb/biossíntese , Proteínas Proto-Oncogênicas c-myb/genética , RNA/genética
17.
Biomaterials ; 172: 30-40, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29715593

RESUMO

Native and engineered tissue development are regulated by the integrative effects of multiple microenvironmental stimuli. Microfabricated bioreactor array platforms can efficiently dissect cue-response networks, and have recently integrated critical 2D and 3D mechanical stimulation for greater physiological relevance. However, a limitation of these approaches is that assessment of tissue functional properties is typically limited to end-point analyses. Here we report a new deformable membrane platform with integrated strain sensors that enables mechanical stretching or compression of 3D cell-hydrogel arrays and simultaneous measurement of hydrogel construct stiffness in situ. We tested the ability of the integrated strain sensors to measure the evolution of the stiffness of cell-hydrogel constructs for two cases. First, we demonstrated in situ stiffness monitoring of degradable poly (ethylene glycol)-norbornene (PEG-NB) hydrogels embedded with mesenchymal stromal cells (MSCs) and cultured with or without cyclic tensile stimulation for up to 15 days. Whereas statically-cultured hydrogels degraded and softened throughout the culture period, mechanically-stimulated gels initially softened and then recovered their stiffness corresponding to extensive cell network and collagen production. Second, we demonstrated in situ measurement of compressive stiffening of MSC-seeded PEG-NB gels cultured statically under osteogenic conditions, corresponding to increased mineralization and cellularization. This measurement technique can be generalized to other relevant bioreactor and organ-on-a-chip platforms to facilitate online, non-invasive, and high-throughput functional analysis, and to provide insights into the dynamics of engineered tissue development that are otherwise not available.


Assuntos
Ensaios de Triagem em Larga Escala/instrumentação , Hidrogéis/química , Teste de Materiais/métodos , Alicerces Teciduais/química , Adesão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Força Compressiva/efeitos dos fármacos , Humanos , Membranas Artificiais , Células-Tronco Mesenquimais/metabolismo , Microtecnologia/métodos , Norbornanos/química , Polietilenoglicóis/química , Engenharia Tecidual/métodos
18.
Circ Res ; 122(3): 405-416, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29273600

RESUMO

RATIONALE: Aortic valve disease is a cell-mediated process without effective pharmacotherapy. CNP (C-type natriuretic peptide) inhibits myofibrogenesis and osteogenesis of cultured valve interstitial cells and is downregulated in stenotic aortic valves. However, it is unknown whether CNP signaling regulates aortic valve health in vivo. OBJECTIVE: The aim of this study is to determine whether a deficient CNP signaling axis in mice causes accelerated progression of aortic valve disease. METHODS AND RESULTS: In cultured porcine valve interstitial cells, CNP inhibited pathological differentiation via the guanylate cyclase NPR2 (natriuretic peptide receptor 2) and not the G-protein-coupled clearance receptor NPR3 (natriuretic peptide receptor 3). We used Npr2+/- and Npr2+/-;Ldlr-/- mice and wild-type littermate controls to examine the valvular effects of deficient CNP/NPR2 signaling in vivo, in the context of both moderate and advanced aortic valve disease. Myofibrogenesis in cultured Npr2+/- fibroblasts was insensitive to CNP treatment, whereas aged Npr2+/- and Npr2+/-;Ldlr-/- mice developed cardiac dysfunction and ventricular fibrosis. Aortic valve function was significantly impaired in Npr2+/- and Npr2+/-;Ldlr-/- mice versus wild-type littermates, with increased valve thickening, myofibrogenesis, osteogenesis, proteoglycan synthesis, collagen accumulation, and calcification. 9.4% of mice heterozygous for Npr2 had congenital bicuspid aortic valves, with worse aortic valve function, fibrosis, and calcification than those Npr2+/- with typical tricuspid aortic valves or all wild-type littermate controls. Moreover, cGK (cGMP-dependent protein kinase) activity was downregulated in Npr2+/- valves, and CNP triggered synthesis of cGMP and activation of cGK1 (cGMP-dependent protein kinase 1) in cultured porcine valve interstitial cells. Finally, aged Npr2+/-;Ldlr-/- mice developed dilatation of the ascending aortic, with greater aneurysmal progression in Npr2+/- mice with bicuspid aortic valves than those with tricuspid valves. CONCLUSIONS: Our data establish CNP/NPR2 signaling as a novel regulator of aortic valve development and disease and elucidate the therapeutic potential of targeting this pathway to arrest disease progression.


Assuntos
Aneurisma Aórtico/genética , Valva Aórtica/anormalidades , Doenças das Valvas Cardíacas/genética , Peptídeo Natriurético Tipo C/fisiologia , Receptores do Fator Natriurético Atrial/deficiência , Disfunção Ventricular Esquerda/genética , Animais , Aorta/patologia , Aneurisma Aórtico/fisiopatologia , Valva Aórtica/fisiopatologia , Estenose da Valva Aórtica/genética , Estenose da Valva Aórtica/fisiopatologia , Doença da Válvula Aórtica Bicúspide , Calcinose/genética , Calcinose/fisiopatologia , Células Cultivadas , Colágeno/biossíntese , GMP Cíclico/fisiologia , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Matriz Extracelular/patologia , Hiperlipidemias/complicações , Hiperlipidemias/genética , Camundongos , Camundongos Knockout , Miofibroblastos/citologia , Peptídeo Natriurético Tipo C/farmacologia , Osteogênese , Proteoglicanas/biossíntese , Receptores do Fator Natriurético Atrial/fisiologia , Receptores de LDL/deficiência , Receptores de LDL/genética , Suínos , Disfunção Ventricular Esquerda/fisiopatologia
19.
Biomaterials ; 145: 9-22, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28843066

RESUMO

Vascular smooth muscle cells (VSMCs) play essential roles in regulating blood vessel form and function. Regeneration of functional vascular smooth muscle tissue to repair vascular diseases is an area of intense research in tissue engineering and regenerative medicine. For functional vascular smooth muscle tissue regeneration to become a practical therapy over the next decade, the field will need to have access to VSMC sources that are effective, robust and safe. While pluripotent stem cells hold good future promise to this end, more immediate translation is expected to come from approaches that generate functional VSMCs from adult sources of multipotent adipose-derived and bone marrow-derived mesenchymal stromal cells (ASCs and BMSCs). The research to this end is extensive and is dominated by studies relating to classical biochemical signalling molecules used to induce differentiation of ASCs and BMSCs. However, prolonged use of the biochemical induction factors is costly and can cause potential endotoxin contamination in the culture. Over recent years several non-traditional differentiation approaches have been devised to mimic defined aspects of the native micro-environment in which VSMCs reside to contribute to the differentiation of VSMC-like cells from ASCs and BMSCs. In this review, the promises and limitations of several non-traditional culture approaches (e.g., co-culture, biomechanical, and biomaterial stimuli) targeting VSMC differentiation are discussed. The extensive crosstalk between the underlying signalling cascades are delineated and put into a translational context. It is expected that this review will not only provide significant insight into VSMC differentiation strategies for vascular smooth muscle tissue engineering applications, but will also highlight the fundamental importance of engineering the cellular microenvironment on multiple scales (with consideration of different combinatorial pathways) in order to direct cell differentiation fate and obtain cells of a desired and stable phenotype. These strategies may ultimately be applied to different sources of stem cells in the future for a range of biomaterial and tissue engineering disciplines.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Animais , Materiais Biocompatíveis/farmacologia , Fenômenos Biomecânicos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos
20.
Acta Biomater ; 58: 34-43, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28532900

RESUMO

The physical and chemical properties of a biomaterial integrate with soluble cues in the cell microenvironment to direct cell fate and function. Predictable biomaterial-based control of integrated cell responses has been investigated with two-dimensional (2D) screening platforms, but integrated responses in 3D have largely not been explored systematically. To address this need, we developed a screening platform using polyethylene glycol norbornene (PEG-NB) as a model biomaterial with which the polymer wt% (to control elastic modulus) and adhesion peptide types (RGD, DGEA, YIGSR) and densities could be controlled independently and combinatorially in arrays of 3D hydrogels. We applied this platform and regression modeling to identify combinations of biomaterial and soluble biochemical (TGF-ß1) factors that best promoted myofibrogenesis of human mesenchymal stromal cells (hMSCs) in order to inform our understanding of regenerative processes for heart valve tissue engineering. In contrast to 2D culture, our screens revealed that soft hydrogels (low PEG-NB wt%) best promoted spread myofibroblastic cells that expressed high levels of α-smooth muscle actin (α-SMA) and collagen type I. High concentrations of RGD enhanced α-SMA expression in the presence of TGF-ß1 and cell spreading regardless of whether TGF-ß1 was in the culture medium. Strikingly, combinations of peptides that maximized collagen expression depended on the presence or absence of TGF-ß1, indicating that biomaterial properties can modulate MSC response to soluble signals. This combination of a 3D biomaterial array screening platform with statistical modeling is broadly applicable to systematically identify combinations of biomaterial and microenvironmental conditions that optimally guide cell responses. STATEMENT OF SIGNIFICANCE: We present a novel screening platform and methodology to model and identify how combinations of biomaterial and microenvironmental conditions guide cell phenotypes in 3D. Our approach to systematically identify complex relationships between microenvironmental cues and cell responses enables greater predictive power over cell fate in conditions with interacting material design factors. We demonstrate that this approach not only predicts that mesenchymal stromal cell (MSC) myofibrogenesis is promoted by soft, porous 3D biomaterials, but also generated new insights which demonstrate how biomaterial properties can differentially modulate MSC response to soluble signals. An additional benefit of the process includes utilizing both parametric and non parametric analyses which can demonstrate dominant significant trends as well as subtle interactions between biochemical and biomaterial cues.


Assuntos
Valvas Cardíacas , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Norbornanos/química , Peptídeos/química , Polietilenoglicóis/química , Engenharia Tecidual , Humanos , Células-Tronco Mesenquimais/citologia , Fator de Crescimento Transformador beta1/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA