Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Int J Mol Sci ; 24(22)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-38003736

RESUMO

We previously developed several successful decellularization strategies that yielded porcine cardiac extracellular matrices (pcECMs) exhibiting tissue-specific bioactivity and bioinductive capacity when cultured with various pluripotent and multipotent stem cells. Here, we study the tissue-specific effects of the pcECM on seeded human mesenchymal stem cell (hMSC) phenotypes using reverse transcribed quantitative polymerase chain reaction (RT-qPCR) arrays for cardiovascular related gene expression. We further corroborated interesting findings at the protein level (flow cytometry and immunological stains) as well as bioinformatically using several mRNA sequencing and protein databases of normal and pathologic adult and embryonic (organogenesis stage) tissue expression. We discovered that upon the seeding of hMSCs on the pcECM, they displayed a partial mesenchymal-to-epithelial transition (MET) toward endothelial phenotypes (CD31+) and morphologies, which were preceded by an early spike (~Day 3 onward after seeding) in HAND2 expression at both the mRNA and protein levels compared to that in plate controls. The CRISPR-Cas9 knockout (KO) of HAND2 and its associated antisense long non-coding RNA (HAND2-AS1) regulatory region resulted in proliferation arrest, hypertrophy, and senescent-like morphology. Bioinformatic analyses revealed that HAND2 and HAND2-AS1 are highly correlated in expression and are expressed in many different tissue types albeit at distinct yet tightly regulated expression levels. Deviation (downregulation or upregulation) from these basal tissue expression levels is associated with a long list of pathologies. We thus suggest that HAND2 expression levels may possibly fine-tune hMSCs' plasticity through affecting senescence and mesenchymal-to-epithelial transition states, through yet unknown mechanisms. Targeting this pathway may open up a promising new therapeutic approach for a wide range of diseases, including cancer, degenerative disorders, and aging. Nevertheless, further investigation is required to validate these findings and better understand the molecular players involved, potential inducers and inhibitors of this pathway, and eventually potential therapeutic applications.


Assuntos
Células-Tronco Mesenquimais , MicroRNAs , RNA Longo não Codificante , Adulto , Humanos , Animais , Suínos , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Regulação para Baixo , Fatores de Transcrição/metabolismo , RNA Mensageiro , Células-Tronco Mesenquimais/metabolismo , RNA Longo não Codificante/genética , Proliferação de Células/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Transição Epitelial-Mesenquimal , MicroRNAs/genética
2.
Langmuir ; 37(15): 4639-4646, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33826341

RESUMO

We report the stress-strain effect of a stretchable natural rubber (NR)-calcium phosphate composite on the surface wettability (SW) using an innovative approach coupling a uniaxial tensile micromachine, goniometer, and microscope. In situ contact angle measurements in real time were performed during mechanical tension. Our results show that SW is guided by the stress-strain relationship with two different characteristics, depending on the static or dynamic experiments. The results evidenced the limits of the classical theory of wetting. Furthermore, based on the mechanically tunable SW of the system associated with the cytocompatibility of the NR composite, we have modeled such a system for application as a cell support. From the experimental surface energy value, our proposed 3D modeling numerical simulation predicted a window of opportunities for cell-NR survival under mechanical stimuli. The presented data and the thermodynamics-based theoretical approach enable not only accurate correlation of SW with mechanical properties of the NR composite but also provide huge potential for future cell supportability in view of tissue engineering.

3.
J Mater Sci Mater Med ; 24(8): 2013-27, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23670603

RESUMO

Mechanical properties of collagen films are less than ideal for biomaterial development towards musculoskeletal repair or cardiovascular applications. Herein, we present a collagen-cellulose composite film (CCCF) compared against swine small intestine submucosa in regards to mechanical properties, cell growth, and histological analysis. CCCF was additionally characterized by FE-SEM, NMR, mass spectrometry, and Raman Microscopy to elucidate its physical structure, collagen-cellulose composition, and structure activity relationships. Mechanical properties of the CCCF were tested in both wet and dry environments, with anisotropic stress-strain curves that mimicked soft-tissue. Mesenchymal stem cells, human umbilical vein endothelial cells, and human coronary artery smooth muscle cells were able to proliferate on the collagen films with specific cell orientation. Mesenchymal stem cells had a higher proliferation index and were able to infiltrate CCCF to a higher degree than small intestine submucosa. With the underlying biological properties, we present a collagen-cellulose composite film towards forthcoming biomaterial-related applications.


Assuntos
Celulose/química , Colágeno/química , Tecido Conjuntivo , Membranas Artificiais , Células-Tronco Mesenquimais/fisiologia , Animais , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Polaridade Celular/efeitos dos fármacos , Polaridade Celular/fisiologia , Células Cultivadas , Celulose/farmacologia , Colágeno/farmacologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Nanofibras/química , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Alicerces Teciduais/química
4.
ACS Appl Bio Mater ; 3(8): 4974-4986, 2020 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-35021675

RESUMO

The basic requirement of any engineered scaffold is to mimic the native tissue extracellular matrix (ECM). Despite substantial strides in understanding the ECM, scaffold fabrication processes of sufficient product robustness and bioactivity require further investigation, owing to the complexity of the natural ECM. A promising bioacive platform for cardiac tissue engineering is that of decellularized porcine cardiac ECM (pcECM, used here as a soft tissue representative model). However, this platform's complexity and batch-to-batch variability serve as processing limitations in attaining a robust and tunable cardiac tissue-specific bioactive scaffold. To address these issues, we fabricated 3D composite scaffolds (3DCSs) that demonstrate comparable physical and biochemical properties to the natural pcECM using wet electrospinning and functionalization with a pcECM hydrogel. The fabricated 3DCSs are non-immunogenic in vitro and support human mesenchymal stem cells' proliferation. Most importantly, the 3DCSs demonstrate tissue-specific bioactivity in inducing spontaneous cardiac lineage differentiation in human induced pluripotent stem cells (hiPSC) and further support the viability, functionality, and maturation of hiPSC-derived cardiomyocytes. Overall, this work illustrates the technology to fabricate robust yet tunable 3D scaffolds of tissue-specific bioactivity (with a proof of concept provided for cardiac tissues) as a platform for basic materials science studies and possible future R&D application in regenerative medicine.

5.
Biofabrication ; 10(2): 025003, 2018 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-29235444

RESUMO

Here, we have developed a 3D bioprinted microchanneled gelatin hydrogel that promotes human mesenchymal stem cell (hMSC) myocardial commitment and supports native cardiomyocytes (CMs) contractile functionality. Firstly, we studied the effect of bioprinted microchanneled hydrogel on the alignment, elongation, and differentiation of hMSC. Notably, the cells displayed well defined F-actin anisotropy and elongated morphology on the microchanneled hydrogel, hence showing the effects of topographical control over cell behavior. Furthermore, the aligned stem cells showed myocardial lineage commitment, as detected using mature cardiac markers. The fluorescence-activated cell sorting analysis also confirmed a significant increase in the commitment towards myocardial tissue lineage. Moreover, seeded CMs were found to be more aligned and demonstrated synchronized beating on microchanneled hydrogel as compared to the unpatterned hydrogel. Overall, our study proved that microchanneled hydrogel scaffold produced by 3D bioprinting induces myocardial differentiation of stem cells as well as supports CMs growth and contractility. Applications of this approach may be beneficial for generating in vitro cardiac model systems to physiological and cardiotoxicity studies as well as in vivo generating custom designed cell impregnated constructs for tissue engineering and regenerative medicine applications.


Assuntos
Bioimpressão/métodos , Miocárdio/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Diferenciação Celular , Células Cultivadas , Humanos , Hidrogéis , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/citologia , Impressão Tridimensional , Ratos
6.
Sci Rep ; 8(1): 3937, 2018 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-29500447

RESUMO

Tissue development, regeneration, or de-novo tissue engineering in-vitro, are based on reciprocal cell-niche interactions. Early tissue formation mechanisms, however, remain largely unknown given complex in-vivo multifactoriality, and limited tools to effectively characterize and correlate specific micro-scaled bio-mechanical interplay. We developed a unique model system, based on decellularized porcine cardiac extracellular matrices (pcECMs)-as representative natural soft-tissue biomaterial-to study a spectrum of common cell-niche interactions. Model monocultures and 1:1 co-cultures on the pcECM of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) were mechano-biologically characterized using macro- (Instron), and micro- (AFM) mechanical testing, histology, SEM and molecular biology aspects using RT-PCR arrays. The obtained data was analyzed using developed statistics, principal component and gene-set analyses tools. Our results indicated biomechanical cell-type dependency, bi-modal elasticity distributions at the micron cell-ECM interaction level, and corresponding differing gene expression profiles. We further show that hMSCs remodel the ECM, HUVECs enable ECM tissue-specific recognition, and their co-cultures synergistically contribute to tissue integration-mimicking conserved developmental pathways. We also suggest novel quantifiable measures as indicators of tissue assembly and integration. This work may benefit basic and translational research in materials science, developmental biology, tissue engineering, regenerative medicine and cancer biomechanics.


Assuntos
Linhagem da Célula , Fenômenos Biomecânicos , Diferenciação Celular , Técnicas de Cocultura , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Microscopia de Fluorescência , Engenharia Tecidual/métodos
7.
Tissue Eng Part A ; 23(1-2): 69-79, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27784199

RESUMO

Effective cellularization is a key approach to prevent small-caliber (<4 mm) tissue-engineered vascular graft (TEVG) failure and maintain patency and contractility following implantation. To achieve this goal, however, improved biomimicking designs and/or relatively long production times (typically several months) are required. We previously reported on porcine carotid artery decellularization yielding biomechanically stable and cell supportive small-caliber (3-4 mm diameter, 5 cm long) arterial extracellular matrix (scaECM) vascular grafts. In this study, we aimed to study the scaECM graft patency in vivo and possibly improve that patency by graft pre-endothelialization with the recipient porcine autologous cells using our previously reported custom-designed dynamic perfusion bioreactor system. Decellularized scaECM vascular grafts were histologically characterized, their immunoreactivity studied in vitro, and their biocompatibility profile evaluated as a xenograft subcutaneous implantation in a mouse model. To study the scaECM cell support and remodeling ability, pig autologous endothelial and smooth muscle cells (SMCs) were seeded and dynamically cultivated within the scaECM lumen and externa/media, respectively. Finally, endothelialized-only scaECMs-hypothesized as a prerequisite for maintaining graft patency and controlling intimal hyperplasia-were transplanted as an interposition carotid artery graft in a porcine model. Graft patency was evaluated through angiography online and endpoint pathological assessment for up to 6 weeks. Our results demonstrate the scaECM-TEVG biocompatibility preserving a structurally and mechanically stable vascular wall not just following decellularization and recellularization but also after implantation. Using our dynamic perfusion bioreactor, we successfully demonstrated the ability of this TEVG to support in vitro recellularization and remodeling by primary autologous endothelial and SMCs, which were seeded on the lumen and the externa/media layers, respectively. Following transplantation, dynamically endothelialized scaECM-TEVGs remained patent for 6 weeks in a pig carotid interposition bypass model. When compared with nonrevitalized control grafts, reendothelialized grafts provided excellent antithrombogenic activity, inhibited intimal hyperplasia formation, and encouraged media wall infiltration and reorganization with recruited host SMCs. We thus demonstrate that readily available decellularized scaECM can be promptly revitalized with autologous cells in a 3-week period before implantation, indicating applicability as a future platform for vascular reconstructive procedures.


Assuntos
Implante de Prótese Vascular , Prótese Vascular , Artérias Carótidas/cirurgia , Matriz Extracelular , Animais , Autoenxertos , Bioprótese , Artérias Carótidas/fisiopatologia , Camundongos , Suínos
8.
Biomater Sci ; 5(6): 1183-1194, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28513656

RESUMO

Various extracellular matrix (ECM) scaffolds, isolated through decellularization, were suggested as ideal biomimetic materials for 'Functional tissue engineering' (FTE). The decellularization process comprises a compromise between damaging and preserving the ultrastructure and composition of ECM-previously shown to affect cell survival, proliferation, migration, organization, differentiation and maturation. Inversely, the effects of cells on the ECM constructs' biophysical properties, under physiological-like conditions, remain still largely unknown. We hypothesized that by re-cellularizing porcine cardiac ECM (pcECM, as a model scaffold) some of the original biophysical properties of the myocardial tissue can be restored, which are related to the scaffold's surface and the bulk modifications consequent to cellularization. We performed a systematic biophysical assessment of pcECM scaffolds seeded with human mesenchymal stem cells (MSCs), a common multipotent cell source in cardiac regenerative medicine. We report a new type of FTE study in which cell interactions with a composite-scaffold were evaluated from the perspective of their contribution to the biophysical properties of the construct surface (FTIR, WETSEM™) and bulk (DSC, TGA, and mechanical testing). The results obtained were compared with acellular pcECM and native ventricular tissue serving as negative and positive controls, respectively. MSC recellularization resulted in an inter-fiber plasticization effect, increased protein density, masking of acylated glycosaminoglycans (GAGs) and active pcECM remodelling which further stabilized the reseeded construct and increased its denaturation resistance. The systematic approach presented herein, therefore, identifies cells as "biological plasticizers" and yields important methodologies, understanding, and data serving both as a reference as well as possible 'design criteria' for future studies in FTE.


Assuntos
Matriz Extracelular/química , Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Glicosaminoglicanos/química , Humanos , Miocárdio/química , Suínos , Resistência à Tração
9.
Acta Biomater ; 50: 220-233, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27956366

RESUMO

Injectable scaffolds for cardiac tissue regeneration are a promising therapeutic approach for progressive heart failure following myocardial infarction (MI). Their major advantage lies in their delivery modality that is considered minimally invasive due to their direct injection into the myocardium. Biomaterials comprising such scaffolds should mimic the cardiac tissue in terms of composition, structure, mechanical support, and most importantly, bioactivity. Nonetheless, natural biomaterial-based gels may suffer from limited mechanical strength, which often fail to provide the long-term support required by the heart for contraction and relaxation. Here we present newly-developed injectable scaffolds, which are based on solubilized decellularized porcine cardiac extracellular matrix (pcECM) cross-linked with genipin alone or engineered with different amounts of chitosan to better control the gel's mechanical properties while still leveraging the ECM biological activity. We demonstrate that these new biohybrid materials are naturally remodeled by mesenchymal stem cells, while supporting high viabilities and affecting cell morphology and organization. They exhibit neither in vitro nor in vivo immunogenicity. Most importantly, their application in treating acute and long term chronic MI in rat models clearly demonstrates the significant therapeutic potential of these gels in the long-term (12weeks post MI). The pcECM-based gels enable not only preservation, but also improvement in cardiac function eight weeks post treatment, as measured using echocardiography as well as hemodynamics. Infiltration of progenitor cells into the gels highlights the possible biological remodeling properties of the ECM-based platform. STATEMENT OF SIGNIFICANCE: This work describes the development of new injectable scaffolds for cardiac tissue regeneration that are based on solubilized porcine cardiac extracellular matrix (ECM), combined with natural biomaterials: genipin, and chitosan. The design of such scaffolds aims at leveraging the natural bioactivity and unique structure of cardiac ECM, while overcoming its limited mechanical strength, which may fail to provide the long-term support required for heart contraction and relaxation. Here, we present a biocompatible gel-platform with custom-tailored mechanical properties that significantly improve cardiac function when injected into rat hearts following acute and chronic myocardial infarction. We clearly demonstrate the substantial therapeutic potential of these scaffolds, which not only preserved heart functions but also alleviated MI damage, even after the formation of a mature scar tissue.


Assuntos
Matriz Extracelular/química , Hidrogéis , Infarto do Miocárdio/terapia , Miocárdio/metabolismo , Alicerces Teciduais/química , Animais , Linhagem Celular , Quitosana/química , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Iridoides/química , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Ratos , Ratos Wistar
10.
Acta Biomater ; 44: 209-220, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27545814

RESUMO

OBJECTIVE: To evaluate the regenerative capacity of non-supplemented and bioactive patches made of decellularized porcine cardiac extracellular matrix (pcECM) and characterize the biological key factors involved in possible cardiac function (CF) restoration following acute and 8weeks chronic MI. BACKGROUND: pcECM is a key natural biomaterial that can affect cardiac regeneration following myocardial infarction (MI), through mechanisms, which are still not clearly understood. METHODS: Wistar rats underwent MI and received pcECM patch (pcECM-P) treatment in either acute or chronic inflammatory phases. Treated, sham operated (no MI), and control (MI without treatment) animals, were compared through echocardiography, hemodynamics, pathological evaluation and analyses of various mRNA and protein level markers. RESULTS: Our results show that in both acute and long-term chronic MI models, pcECM promotes significant cardiac function improvement, which is correlated to progenitor (GATA4(+), c-kit(+)) and myocyte (MYLC(+), TRPI(+)) recruitment. Interestingly, recruited progenitors, isolated using laser capture microdissection (LCM), expressed both early and late cardiomyocyte (CM) differentiation markers, suggesting differentiation towards the CM lineage. Recruited CM-like cells organized in a partially striated and immature muscle fiber arrangement that presented connexin43 -a crucial mediator of cardiac electrical conductivity. Concomitantly, pcECM was rapidly vascularized, and induced a constructive remodeling process as indicated by increased M2/M1 macrophage phenotypic ratio and pathological evaluation. CONCLUSIONS: Acellular pcECM patch implants alone, i.e., without added biologics, are bioactive, and exert potent efficacy, stimulating biological regenerative processes that cooperatively lead to a cardiac progenitor-based restoration of function, even after scar tissue had already formed. STATEMENT OF SIGNIFICANCE: MI ('heart attack') remains the leading cause of heart failure and death in developed-countries. Restoration of cardiac function requires active turnover of damaged heart contracting cells (CM), however, CM endogenous regeneration is not efficient and is a matter of controversy. We show that a bioactive biomaterial alone-decellularized heart tissue (pcECM)-without added cells or growth factors, can elicit a complex regenerative response even after irreversible scarring. The pcECM patch induces macrophage polarization towards constructive remodeling and cardiomyocyte progenitor cell (GATA4(+), c-kit(+)) recruitment (evidenced at both mRNA and protein levels) resulting in de novo immature striated-like muscle patterns (MLC(+), TrpI(+), connexin43(+)). We, therefore, suggest this bioactive pcECM can model cardiac regeneration, and serve as a candidate for fast-track clinical application.


Assuntos
Cicatriz/patologia , Matriz Extracelular/metabolismo , Miocárdio/metabolismo , Regeneração , Células-Tronco/citologia , Animais , Contagem de Células , Hemodinâmica , Implantes Experimentais , Macrófagos/patologia , Masculino , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Ratos Wistar , Sus scrofa
11.
Tissue Eng Part A ; 21(9-10): 1507-19, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25602926

RESUMO

Functional vascularization is a prerequisite for cardiac tissue engineering of constructs with physiological thicknesses. We previously reported the successful preservation of main vascular conduits in isolated thick acellular porcine cardiac ventricular ECM (pcECM). We now unveil this scaffold's potential in supporting human cardiomyocytes and promoting new blood vessel development ex vivo, providing long-term cell support in the construct bulk. A custom-designed perfusion bioreactor was developed to remodel such vascularization ex vivo, demonstrating, for the first time, functional angiogenesis in vitro with various stages of vessel maturation supporting up to 1.7 mm thick constructs. A robust methodology was developed to assess the pcECM maximal cell capacity, which resembled the human heart cell density. Taken together these results demonstrate feasibility of producing physiological-like constructs such as the thick pcECM suggested here as a prospective treatment for end-stage heart failure. Methodologies reported herein may also benefit other tissues, offering a valuable in vitro setting for "thick-tissue" engineering strategies toward large animal in vivo studies.


Assuntos
Matriz Extracelular/metabolismo , Miocárdio/metabolismo , Neovascularização Fisiológica , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Reatores Biológicos , Técnicas de Cocultura , Estudos de Viabilidade , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Miocárdio/citologia , Sus scrofa
12.
Biomatter ; 4: e28238, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24553126

RESUMO

Human umbilical vein endothelial cells (HUVECs) were successfully entrapped in polyethylene oxide (PEO) core /polycaprolactone (PCL) shell electrospun fibers thus creating a "bioactive fiber." The viability and release of biomolecules from the entrapped cells in the bioactive fibers were characterized. A key modification to the core solution was the inclusion of 50% fetal bovine serum (FBS), which improved cell viability substantially. The fluorescein diacetate (FDA) staining revealed that the entrapped cells were intact and viable immediately after the electrospinning process. A long-term cell viability assay using AlamarBlue® showed that cells were viable for over two weeks. Secreted Interleukin-8 (IL-8) was monitored as a candidate released protein, which can also act as an indicator of HUVEC stress. These results demonstrated that HUVECs could be entrapped within the electrospun scaffold with the potential of controllable cell deposition and the creation of a bioactive fibrous scaffold with extended functionality.


Assuntos
Materiais Biocompatíveis/química , Células Endoteliais da Veia Umbilical Humana , Alicerces Teciduais/química , Sobrevivência Celular/efeitos dos fármacos , Células Imobilizadas , Humanos , Interleucina-8/metabolismo , Poliésteres/química , Polietilenoglicóis/química , Porosidade , Soluções/química , Engenharia Tecidual/métodos
13.
Tissue Eng Part A ; 19(9-10): 1155-64, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23216214

RESUMO

In most tissue engineering applications, understanding the factors affecting the growth dynamics of coculture systems is crucial for directing the population toward a desirable regenerative process. Yet, no comprehensive analysis method exists to quantify coculture population dynamics, let alone, a unifying model addressing the "environmental" factors influencing cell growth, all together. Here we suggest a modification of the Lotka-Volterra model to analyze the population dynamics of cocultured cells and predict their growth profiles for tissue engineering applications. This model, commonly used to describe the population dynamics of a prey and predator sharing a closed ecological niche, was found to fit our empirical data on cocultures of endothelial cells (ECs) and mesenchymal stem cells (MSCs) that have been widely investigated for their regenerative potential. Applying this model to cocultures of this sort allows us to quantify the effect that culturing conditions have on the way cell growth is affected by the same cells or by the other cells in the coculture. We found that in most cases, EC growth was inhibited by the same cells but promoted by MSCs. The principles resulting from this analysis can be used in various applications to guide the population toward a desired direction while shedding new light on the fundamental interactions between ECs and MSCs. Similar results were also demonstrated on complex substrates made from decellularized porcine cardiac extracellular matrix, where growth occurred only after coculturing ECs and MSCs together. Finally, this unique implementation of the Lotka-Volterra model may also be regarded as a roadmap for using such models with other potentially regenerative cocultures in various applications.


Assuntos
Células Endoteliais/citologia , Células-Tronco Mesenquimais/citologia , Modelos Teóricos , Sobrevivência Celular , Células Cultivadas , Humanos , Engenharia Tecidual
14.
Tissue Eng Part C Methods ; 19(8): 620-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23265414

RESUMO

The clinical success of tissue-engineered constructs commonly requires mechanical properties that closely mimic those of the human tissue. Determining the viscoelastic properties of such biomaterials and the factors governing their failure profiles, however, has proven challenging, although collecting extensive data regarding their tensile behavior is straightforward. The easily calculated Young's modulus remains the most reported mechanical measure, regardless of its limitations, even though single-relaxation-time (SRT) models can provide much more information, which remain scarce due to a lack of manageable tools for implementing these models. We developed an easy-to-use algorithm for applying the Zener SRT model and determining the elastic moduli, viscosity, and failure profiles of materials under different mechanical tests in a user-independent manner. The algorithm was validated on the data resulting from tensile tests on native and decellularized porcine cardiac tissue, previously suggested as a promising scaffold material for cardiac tissue engineering. This analysis yields new and more accurate measurements such as the elastic moduli and viscosity, the model's relaxation time, and information on the factors governing the materials' failure profiles. These measurements indicate that the viscoelasticity and strength of the decellularized acellular extracellular matrix (ECM) are similar to those of native tissue, although its elasticity and apparent viscosity are higher. Nonetheless, reseeding and culturing the ECM with mesenchymal stem cells was shown to partially restore the mechanical properties lost after decellularization. We propose this algorithm as a platform for soft-tissue analysis that can provide comparable and unbiased measures for characterizing viscoelastic biomaterials commonly used in tissue engineering.


Assuntos
Módulo de Elasticidade , Matriz Extracelular/química , Modelos Biológicos , Miocárdio/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Humanos , Suínos , Viscosidade
15.
Tissue Eng Part A ; 18(3-4): 411-22, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21919798

RESUMO

Patients with small caliber artery disorders, often lack the suitable autologous tissue needed for bypassing diseased vessels or for other vascular reconstructive procedures. We propose to decellularize small caliber porcine carotid artery, then recellularize it with vascular cells and function as scaffold for tissue engineering vascular graft replacements. Based on a modified decellularization method developed in our laboratory, the cellular contents of small caliber (<4 mm) arteries were carefully removed using an enzymatic and detergent decellularization procedure. Decellularization efficiency was evaluated using histology and scanning electron microscopy, which demonstrated the absence of cellular remains in the artery wall. Proteomic analysis of the scaffold revealed that the decellularized vessels retained their major extracellular matrix protein composition. Mechanical analyses revealed no significant change in the extracellular matrix (ECM) properties versus the native artery. The decellularized artery was reseeded with human umbilical vein endothelial cells (HUVECs) and smooth muscle cells (SMCs) and cultured under static or dynamic conditions in a perfusion bioreactor designed and developed in our laboratory for these studies. Dynamic co-culturing of SMC and HUVEC, in this custom-made perfusion bioreactor, led to a higher infiltration, migration and proliferation of SMC toward the media and to a more confluent endothelium formation on the luminal surface when compared with static culturing. In addition, vascular media remodeling by SMC correlated to the expression of remodeling related genes assessed by real-time reverse transcription-polymerase chain reaction and HUVEC cultivation contributed to the remodeling of several basement membrane proteins stained using immunohistochemistry. All together, these findings indicate the potential of such decellularized arterial ECM for future small caliber vascular graft reconstruction therapies.


Assuntos
Prótese Vascular , Artérias Carótidas/anatomia & histologia , Endotélio Vascular/crescimento & desenvolvimento , Matriz Extracelular/metabolismo , Engenharia Tecidual/métodos , Túnica Média/fisiologia , Animais , Fenômenos Biomecânicos , Reatores Biológicos , Sobrevivência Celular , Matriz Extracelular/genética , Matriz Extracelular/ultraestrutura , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Miócitos de Músculo Liso/citologia , Perfusão , Proteômica , Reprodutibilidade dos Testes , Ovinos , Sus scrofa , Técnicas de Cultura de Tecidos , Alicerces Teciduais
16.
Tissue Eng Part A ; 18(19-20): 2125-37, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22663095

RESUMO

The decellularization of porcine heart tissue offers many opportunities for the production of physiologically relevant myocardial mimetic scaffolds. Earlier, we reported the successful isolation of a thin porcine cardiac extracellular matrix (pcECM) exhibiting relevant bio-mechanical properties for myocardial tissue engineering. Nevertheless, since native cardiac tissue is much thicker, such thin scaffolds may offer limited regeneration capacity. However, generation of thicker myocardial mimetic tissue constructs is hindered by diffusion limitations (~100 µm), and the lack of a proper vascular-like network within these constructs. In our present work, we focused on optimizing the decellularization procedure for thicker tissue slabs (10-15 mm), while retaining their inherent vasculature, and on characterizing the resulting pcECM. The trypsin/Triton-based perfusion procedure that resulted in a nonimmunogenic and cell-supportive pcECM was found to be more effective in cell removal and in the preservation of fiber morphology and structural characteristics than stirring, sonication, or sodium dodecyl sulfate/Triton-based procedures. Mass spectroscopy revealed that the pcECM is mainly composed of ECM proteins with no apparent cellular protein remains. Mechanical testing indicated that the obtained pcECM is viscoelastic in nature and possesses the typical stress-strain profile of biological materials. It is stiffer than native tissue yet exhibits matched mechanical properties in terms of energy dissipation, toughness, and ultimate stress behavior. Vascular network functionality was maintained to the first three-four branches from the main coronary vessels. Taken together, these results reaffirm the efficiency of the decellularization procedure reported herein for yielding thick nonimmunogenic cell-supportive pcECM scaffolds, preserving both native tissue ultra-structural properties and an inherent vascular network. When reseeded with the appropriate progenitor cells, these scaffolds can potentially serve as ex vivo screening platforms for new therapeutics, as models for human cardiac ECM, or as biomedical constructs for patch or transmural transplantation strategies.


Assuntos
Matriz Extracelular/química , Miocárdio/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Adesão Celular/fisiologia , Células Cultivadas , Feminino , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Microscopia Eletrônica de Varredura , Ratos , Suínos
17.
Expert Opin Biol Ther ; 11(8): 1055-77, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21542780

RESUMO

INTRODUCTION: Various engineered 'cell-platforms' have been reported in recent years for the possible treatment of myocardial infarction (MI) and end-stage heart failure. These engineered platforms rely on two key factors: cells and/or biomaterial scaffolds for the regeneration of the infarcted heart tissue. AREAS COVERED: Two major cell-platform approaches are described and broadly categorized as 'injectable cell platforms' and 'patch-based cell platforms'. The recent advancements in these cell-platforms in terms of their relative successes in-vivo as well as their clinical feasibility are summarized. Natural as well as synthetic scaffolds, with or without the cellular component, are compared with cell based therapy alone. This review focuses on achievements, as well as the gaps that are presently checking any progress towards producing clinically relevant panacea for myocardial regeneration. EXPERT OPINION: Cardiac and induced pluripotent stem cells will probably be the focus of future research. The combined cell-biomaterial scaffold therapy is superior to cell therapy alone. Nevertheless, encouraging pre-clinical successes have limited translation into clinical practice due to limited cell survival post transplantation, inadequate construct thicknesses for human-sized hearts and the traditional use of 'flat (2D) tissue culture' techniques. The development of complementary dynamic 3D cultivation platforms will probably lead to improved outcomes and enable fast screening of various therapeutic approaches.


Assuntos
Doenças Cardiovasculares/terapia , Transplante de Células , Miócitos Cardíacos/fisiologia , Regeneração , Engenharia Tecidual , Animais , Humanos , Miócitos Cardíacos/citologia
18.
Tissue Eng Part C Methods ; 16(4): 671-83, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19780649

RESUMO

We have developed an efficient decellularization process for the isolation of extracellular matrix (ECM) from native cardiac tissue. The isolated ECM exhibited desirable mechanical properties in terms of elasticity, strength and durability-properties required from scaffolds used for cardiac tissue repair. This study further investigates the potential use of this scaffold for cardiac tissue engineering in terms of interactions with seeded cells and biocompatibility. We used the commonly studied fibroblasts, cardiomyocytes, and mesenchymal stem cells, which were isolated and seeded onto the scaffold. Cell density and distribution were followed by 3,3'-dioctadecyloxacarbocyanine perchlorate staining, and their proliferation and viability were assessed by AlamarBlue assay and fluorecein-diacetate/propidium iodide staining. Fibroblast-seeded scaffolds shrank to 1-2 mm(3) spheroids, and their glycosaminoglycans significantly increased by 23%. The expression of ECM remodeling-related mRNAs of collagens I and III, matrix metalloproteinase 2, and type 1 tissue inhibitor of metalloproteinases was quantified by real-time polymerase chain reaction, and was found significantly elevated in fibroblast-seeded scaffold, compared with the control cells on plates. Fibroblast-seeded scaffolds lost some flexibility, yet gained strength compared with the acellular scaffolds, as shown by mechanical testing. Scaffold seeded with cardiomyocyte began to beat in concert few days after seeding, and the myocytes expressed typical functional cardiac markers such as alpha-actinin, troponin I, and connexin43. The cells revealed aligned elongated morphology, as presented by immunofluorescent staining and scanning electron microscopy. Mesenchymal stem cell-seeded scaffolds maintained viability over 24 days in culture. These findings further strengthen the potential use of acellular cardiac ECM as a biomaterial for heart regeneration.


Assuntos
Materiais Biocompatíveis/farmacologia , Matriz Extracelular/metabolismo , Fibroblastos/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Bioensaio , Fenômenos Biomecânicos/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colágeno/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/ultraestrutura , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Imuno-Histoquímica , Imunofenotipagem , Ativação de Macrófagos/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Contração Miocárdica/efeitos dos fármacos , Miocárdio/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Reprodutibilidade dos Testes , Ovinos , Sus scrofa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA