Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 12(13)2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37443731

RESUMO

Heart regeneration after myocardial infarction (MI) using human stem cell-derived cardiomyocytes (CMs) is rapidly accelerating with large animal and human clinical trials. However, vascularization methods to support the engraftment, survival, and development of implanted CMs in the ischemic environment of the infarcted heart remain a key and timely challenge. To this end, we developed a dual remuscularization-revascularization therapy that is evaluated in a rat model of ischemia-reperfusion MI. This study details the differentiation of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) for engineering cardiac tissue containing patterned engineered vessels 400 µm in diameter. Vascularized engineered human myocardial tissues (vEHMs) are cultured in static conditions or perfused in vitro prior to implantation and evaluated after two weeks. Immunohistochemical staining indicates improved engraftment of hiPSC-CMs in in vitro-perfused vEHMs with greater expression of SMA+ vessels and evidence of inosculation. Three-dimensional vascular reconstructions reveal less tortuous and larger intra-implant vessels, as well as an improved branching hierarchy in in vitro-perfused vEHMs relative to non-perfused controls. Exploratory RNA sequencing of explanted vEHMs supports the hypothesis that co-revascularization impacts hiPSC-CM development in vivo. Our approach provides a strong foundation to enhance vEHM integration, develop hierarchical vascular perfusion, and maximize hiPSC-CM engraftment for future regenerative therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Infarto do Miocárdio , Humanos , Ratos , Animais , Arteríolas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Infarto do Miocárdio/terapia , Infarto do Miocárdio/metabolismo , Perfusão
2.
Bioengineering (Basel) ; 10(5)2023 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-37237658

RESUMO

Despite the overwhelming use of cellularized therapeutics in cardiac regenerative engineering, approaches to biomanufacture engineered cardiac tissues (ECTs) at clinical scale remain limited. This study aims to evaluate the impact of critical biomanufacturing decisions-namely cell dose, hydrogel composition, and size-on ECT formation and function-through the lens of clinical translation. ECTs were fabricated by mixing human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) and human cardiac fibroblasts into a collagen hydrogel to engineer meso-(3 × 9 mm), macro- (8 × 12 mm), and mega-ECTs (65 × 75 mm). Meso-ECTs exhibited a hiPSC-CM dose-dependent response in structure and mechanics, with high-density ECTs displaying reduced elastic modulus, collagen organization, prestrain development, and active stress generation. Scaling up, cell-dense macro-ECTs were able to follow point stimulation pacing without arrhythmogenesis. Finally, we successfully fabricated a mega-ECT at clinical scale containing 1 billion hiPSC-CMs for implantation in a swine model of chronic myocardial ischemia to demonstrate the technical feasibility of biomanufacturing, surgical implantation, and engraftment. Through this iterative process, we define the impact of manufacturing variables on ECT formation and function as well as identify challenges that must still be overcome to successfully accelerate ECT clinical translation.

3.
PLoS One ; 18(2): e0280406, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36745602

RESUMO

Recent advances in human induced pluripotent stem cell (hiPSC)-derived cardiac microtissues provide a unique opportunity for cardiotoxic assessment of pharmaceutical and environmental compounds. Here, we developed a series of automated data processing algorithms to assess changes in action potential (AP) properties for cardiotoxicity testing in 3D engineered cardiac microtissues generated from hiPSC-derived cardiomyocytes (hiPSC-CMs). Purified hiPSC-CMs were mixed with 5-25% human cardiac fibroblasts (hCFs) under scaffold-free conditions and allowed to self-assemble into 3D spherical microtissues in 35-microwell agarose gels. Optical mapping was performed to quantify electrophysiological changes. To increase throughput, AP traces from 4x4 cardiac microtissues were simultaneously acquired with a voltage sensitive dye and a CMOS camera. Individual microtissues showing APs were identified using automated thresholding after Fourier transforming traces. An asymmetric least squares method was used to correct non-uniform background and baseline drift, and the fluorescence was normalized (ΔF/F0). Bilateral filtering was applied to preserve the sharpness of the AP upstroke. AP shape changes under selective ion channel block were characterized using AP metrics including stimulation delay, rise time of AP upstroke, APD30, APD50, APD80, APDmxr (maximum rate change of repolarization), and AP triangulation (APDtri = APDmxr-APD50). We also characterized changes in AP metrics under various ion channel block conditions with multi-class logistic regression and feature extraction using principal component analysis of human AP computer simulations. Simulation results were validated experimentally with selective pharmacological ion channel blockers. In conclusion, this simple and robust automated data analysis pipeline for evaluating key AP metrics provides an excellent in vitro cardiotoxicity testing platform for a wide range of environmental and pharmaceutical compounds.


Assuntos
Potenciais de Ação , Cardiotoxicidade , Células-Tronco Pluripotentes Induzidas , Humanos , Potenciais de Ação/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Canais Iônicos , Miócitos Cardíacos/fisiologia
4.
ALTEX ; 40(1): 103-116, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35648122

RESUMO

Environmental factors play a substantial role in determining cardiovascular health, but data informing the risks presented by environmental toxicants is insufficient. In vitro new approach methodologies (NAMs) offer a promising approach with which to address the limitations of traditional in vivo and in vitro assays for assessing cardiotoxicity. Driven largely by the needs of pharmaceutical toxicity testing, considerable progress in developing NAMs for cardiotoxicity analysis has already been made. As the scientific and regulatory interest in NAMs for environmental chemicals continues to grow, a thorough understanding of the unique features of environmental cardiotoxicants and their associated cardiotoxicities is needed. Here, we review the key characteristics of as well as important regulatory and biological considerations for fit-for-purpose NAMs for environmental cardiotoxicity. By emphasizing the challenges and opportunities presented by NAMs for environmental cardiotoxicity we hope to accelerate their development, acceptance, and application.


Assuntos
Cardiotoxicidade , Células-Tronco Pluripotentes Induzidas , Humanos , Testes de Toxicidade/métodos , Miócitos Cardíacos , Preparações Farmacêuticas
5.
Polymers (Basel) ; 14(21)2022 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-36365565

RESUMO

Myocardial infarction is a leading cause of death worldwide and has severe consequences including irreversible damage to the myocardium, which can lead to heart failure. Cardiac tissue engineering aims to re-engineer the infarcted myocardium using tissues made from human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to regenerate heart muscle and restore contractile function via an implantable epicardial patch. The current limitations of this technology include both biomanufacturing challenges in maintaining tissue integrity during implantation and biological challenges in inducing cell alignment, maturation, and coordinated electromechanical function, which, when overcome, may be able to prevent adverse cardiac remodeling through mechanical support in the injured heart to facilitate regeneration. Polymer scaffolds serve to mechanically reinforce both engineered and host tissues. Here, we introduce a novel biodegradable, customizable scaffold composed of wet-spun polycaprolactone (PCL) microfibers to strengthen engineered tissues and provide an anisotropic mechanical environment to promote engineered tissue formation. We developed a wet-spinning process to produce consistent fibers which are then collected on an automated mandrel that precisely controls the angle of intersection of fibers and their spacing to generate mechanically anisotropic scaffolds. Through optimization of the wet-spinning process, we tuned the fiber diameter to 339 ± 31 µm and 105 ± 9 µm and achieved a high degree of fidelity in the fiber structure within the scaffold (fiber angle within 1.8° of prediction). Through degradation and mechanical testing, we demonstrate the ability to maintain scaffold mechanical integrity as well as tune the mechanical environment of the scaffold through structure (Young's modulus of 120.8 ± 1.90 MPa for 0° scaffolds, 60.34 ± 11.41 MPa for 30° scaffolds, 73.59 ± 3.167 MPa for 60° scaffolds, and 49.31 ± 6.90 MPa for 90° scaffolds), while observing decreased hysteresis in angled vs. parallel scaffolds. Further, we embedded the fibrous PCL scaffolds in a collagen hydrogel mixed with hiPSC-CMs to form engineered cardiac tissue with high cell survival, tissue compaction, and active contractility of the hiPSC-CMs. Through this work, we develop and optimize a versatile biomanufacturing process to generate customizable PCL fibrous scaffolds which can be readily utilized to guide engineered tissue formation and function.

6.
Methods Mol Biol ; 2485: 147-157, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35618904

RESUMO

Risk assessment assays for chemically induced arrhythmia are critical, but significant limitations exist with current cardiotoxicity testing, including a focus on single select ion channels, the use of non-human species in vitro and in vivo, and limited direct physiological translation. To be predictive of actual adverse clinical arrhythmic risk, arrhythmia assessment models for chemicals and drugs should be fit-for-purpose and suited for evaluating compounds in which the mechanism of action may not be entirely known. Here, we describe methods for efficient and reliable screening for arrhythmogenic cardiotoxicity with a 3D human cardiac microtissue model using purified human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and human cardiac fibroblasts. Applying optical mapping of voltage and calcium-sensitive dyes-an established approach to evaluate cardiac action potentials and calcium transients-to 3D heterotypic cardiac myocyte-fibroblast tissues allows for the generation and functional analysis of a large number of individual microtissues to provide greater throughput and high statistical power in analyses. Hundreds of microtissues in standard cell culture plates can be produced with low variability beat-to-beat, microtissue-to-microtissue, and across hiPSC-cardiomyocyte differentiation batches, reducing the number of microtissues required per condition for predictive outputs. The platform described here can be used as a sensitive, efficient, and predictive preclinical model validated for the purpose of assessing human pro-arrhythmic risk.


Assuntos
Cálcio , Miócitos Cardíacos , Arritmias Cardíacas/induzido quimicamente , Cardiotoxicidade , Fibroblastos , Humanos
7.
Stem Cells Transl Med ; 11(1): 97-106, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35641165

RESUMO

Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have profound utility in generating functional human engineered cardiac tissues (ECT) for heart repair. However, the field at large is concerned about the relative immaturity of these hiPSC-CMs as we aim to develop clinically relevant models for regenerative therapy and drug testing. Herein, we develop a novel calcium (Ca2+) conditioning protocol that maintains ECTs in a physiological range of Ca2+ and assesses contractility in increasing calcium environments. Lactate-based selection served as a method to purify and shift the metabolic profile of hiPSC-CMs to evaluate the role of metabolism on Ca2+ sensitivity. After 2 weeks, we observe 2-fold greater peak twitch stress in high-Ca2+ conditioned ECTs, despite having lower stiffness and no change in Ca2+ sensitivity of twitch force. Interestingly, the force-calcium relationship reveals higher Ca2+ sensitivity in lactate conditioned tissues, suggesting that metabolic maturation alters mitochondrial Ca2+ buffering and regulation. Ca2+ sensitivity and force amplitude are not coupled, as lactate conditioned tissues produce force comparable to that of controls in high calcium environments. An upregulation of calcium handling protein gene expression likely contributes to the greater Ca2+ sensitivity in lactate conditioned hiPSC-CMs. Our findings support the use of physiological Ca2+ to enhance the functional maturation of excitation-contraction coupling in hiPSC-CMs and demonstrate that metabolic changes induced by lactate conditioning alter cardiomyocyte sensitivity to external Ca2+. These conditioning methods may be used to advance the development of engineered human cardiac tissue for translational applications in vitro and in vivo as a regenerative therapy.


Assuntos
Células-Tronco Pluripotentes Induzidas , Cálcio/metabolismo , Diferenciação Celular/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lactatos/metabolismo , Miócitos Cardíacos/metabolismo , Engenharia Tecidual/métodos
8.
ACS Appl Bio Mater ; 5(6): 2461-2480, 2022 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-35623101

RESUMO

Despite numerous advances in treatments for cardiovascular disease, heart failure (HF) remains the leading cause of death worldwide. A significant factor contributing to the progression of cardiovascular diseases into HF is the loss of functioning cardiomyocytes. The recent growth in the field of cardiac tissue engineering has the potential to not only reduce the downstream effects of injured tissues on heart function and longevity but also re-engineer cardiac function through regeneration of contractile tissue. One leading strategy to accomplish this is via a cellularized patch that can be surgically implanted onto a diseased heart. A key area of this field is the use of tissue scaffolds to recapitulate the mechanical and structural environment of the native heart and thus promote engineered myocardium contractility and function. While the strong mechanical properties and anisotropic structural organization of the native heart can be largely attributed to a robust extracellular matrix, similar strength and organization has proven to be difficult to achieve in cultured tissues. Polycaprolactone (PCL) is an emerging contender to fill these gaps in fabricating scaffolds that mimic the mechanics and structure of the native heart. In the field of cardiovascular engineering, PCL has recently begun to be studied as a scaffold for regenerating the myocardium due to its facile fabrication, desirable mechanical, chemical, and biocompatible properties, and perhaps most importantly, biodegradability, which make it suitable for regenerating and re-engineering function to the heart after disease or injury. This review focuses on the application of PCL as a scaffold specifically in myocardium repair and regeneration and outlines current fabrication approaches, properties, and possibilities of PCL incorporation into engineered myocardium, as well as provides suggestions for future directions and a roadmap toward clinical translation of this technology.


Assuntos
Poliésteres , Alicerces Teciduais , Miócitos Cardíacos , Poliésteres/química , Regeneração , Alicerces Teciduais/química
9.
Cell Mol Bioeng ; 14(5): 441-457, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34777603

RESUMO

INTRODUCTION: Although atrial fibrillation is the most prevalent disorder of electrical conduction, the mechanisms behind atrial arrhythmias remain elusive. To address this challenge, we developed a robust in vitro model of 3D atrial microtissue from human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and evaluated chamber-specific chemical responses experimentally and computationally. METHODS: We differentiated atrial and ventricular cardiomyocytes (aCMs/vCMs) from GCaMP6f-expressing hiPSCs and assessed spontaneous AP activity using fluorescence imaging. Self-assembling 3D microtissues were formed with lactate purified CMs and 5% human cardiac fibroblasts and electrically stimulated for one week before high resolution action potential (AP) optical mapping. AP responses to the atrial-specific potassium repolarizing current I Kur-blocker 4-Aminopyridine (4-AP) and funny current I f-blocker Ivabradine were characterized within their therapeutic window. Finally, we expanded upon a published hiPSC-CM computational model by incorporating the atrial-specific I Kur current, modifying ion channel conductances to match the AP waveforms of our microtissues, and employing the updated model to reinforce our experimental findings. RESULTS: High purity CMs (> 75% cTnT+) demonstrated subtype specification by MLC2v expression. Spontaneous beating rates significantly decreased following 3D microtissue formation, with atrial microtissues characterized by their faster spontaneous beating rate, slower AP rise time, and shorter AP duration (APD) compared to ventricular microtissues. We measured atrial-specific responses, including dose-dependent APD prolongation with 4-AP treatment and dose-dependent reduction in spontaneous activity post-Ivabradine treatment. CONCLUSION: The presented in vitro platform for screening atrial-specific responses is both robust and sensitive, with high throughput, enabling studies focused at elucidating the mechanisms underlying atrial arrhythmias. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12195-021-00703-x.

10.
J Mech Behav Biomed Mater ; 121: 104627, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34130078

RESUMO

Cardiac patch therapies promise to restore heart function and lower the risk of heart failure after heart attack. Fiber-matrix engineered tissue scaffolds have gained significant attention due to their tunable micro-structures, providing nonlinear mechanical properties similar to native anisotropic heart tissues. Mechanical properties of engineered scaffolds directly affect the stress fields generated inside and around the tissue scaffolds and have significant impact on the tissue functionality. Currently, biomedical cardiac patches are designed through experimentation and there exists a need for an accurate model that will allow micro-structural design optimization and analysis of effectiveness of the implanted patches. We have developed a three-dimensional large strain continuum model that can predict nonlinear, anisotropic mechanical response of engineered tissue scaffolds that have two orientation families of fibers inside a bulk hydrogel matrix. We have validated the predictive capability of our continuum model for the fiber-matrix composite using selected experiments and a suite of detailed finite element analysis that incorporated the micro-structural details of the composites. Comparing the continuum model predictions (1 element) against the representative volume micro-structural geometry finite element simulations (with greater than 4,00,000 elements), we show that the proposed model can accurately predict nonlinear mechanical behavior of highly anisotropic tissue scaffolds in both the longitudinal and transverse directions, as a function of the critical design parameters inter-fiber angle and fiber spacing. We show that the model can also capture native heart tissue's anisotropic large strain mechanical response. We implemented our model in the finite element software Abaqus by writing a user material subroutine UANISOHYPER and demonstrated its predictive abilities by conducting a full three-dimensional analysis of engineered tissue patch application on an infarcted heart.


Assuntos
Coração , Engenharia Tecidual , Anisotropia , Simulação por Computador , Análise de Elementos Finitos , Estresse Mecânico
11.
Sci Rep ; 11(1): 10228, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33986332

RESUMO

Cardiotoxicity of pharmaceutical drugs, industrial chemicals, and environmental toxicants can be severe, even life threatening, which necessitates a thorough evaluation of the human response to chemical compounds. Predicting risks for arrhythmia and sudden cardiac death accurately is critical for defining safety profiles. Currently available approaches have limitations including a focus on single select ion channels, the use of non-human species in vitro and in vivo, and limited direct physiological translation. We have advanced the robustness and reproducibility of in vitro platforms for assessing pro-arrhythmic cardiotoxicity using human induced pluripotent stem cell-derived cardiomyocytes and human cardiac fibroblasts in 3-dimensional microtissues. Using automated algorithms and statistical analyses of eight comprehensive evaluation metrics of cardiac action potentials, we demonstrate that tissue-engineered human cardiac microtissues respond appropriately to physiological stimuli and effectively differentiate between high-risk and low-risk compounds exhibiting blockade of the hERG channel (E4031 and ranolazine, respectively). Further, we show that the environmental endocrine disrupting chemical bisphenol-A (BPA) causes acute and sensitive disruption of human action potentials in the nanomolar range. Thus, this novel human 3D in vitro pro-arrhythmic risk assessment platform addresses critical needs in cardiotoxicity testing for both environmental and pharmaceutical compounds and can be leveraged to establish safe human exposure levels.


Assuntos
Miócitos Cardíacos/efeitos dos fármacos , Medição de Risco/métodos , Engenharia Tecidual/métodos , Potenciais de Ação/efeitos dos fármacos , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Arritmias Cardíacas/prevenção & controle , Cardiotoxicidade/prevenção & controle , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Morte Súbita Cardíaca/prevenção & controle , Fibroblastos/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Modelos Biológicos , Contração Miocárdica/efeitos dos fármacos , Reprodutibilidade dos Testes
12.
J Biomed Mater Res A ; 109(9): 1726-1736, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33733622

RESUMO

A formidable challenge in regenerative medicine is the development of stable microvascular networks to restore adequate blood flow or to sustain graft viability and long-term function in implanted or ischemic tissues. In this work, we develop a biomimetic approach to increase the binding affinity of the extracellular matrix for the class of heparin-binding growth factors to localize and control the release of proangiogenic cues while maintaining their bioactivity. Sulfate and heparin moieties are covalently coupled to alginate, and alginate microspheres are produced and used as local delivery depots for vascular endothelial growth factor (VEGF). Release of VEGF from sulfate-alginate and heparin-alginate bulk hydrogels and microspheres was sustained over 14 days. In vitro evaluation with human induced pluripotent stem cell (hiPSC)-derived endothelial cells and aortic ring assay in a chemically defined hydrogel demonstrates development of primitive three-dimensional vessel-like networks in the presence of VEGF released from the chemically modified alginate microspheres. Furthermore, our results suggest that the sulfate groups available on the chemically modified alginate microspheres promote some new vessel formation even in VEGF-free samples. Based on this evidence, we conclude that sulfate- and heparin-alginate hydrogels are adaptive and bioactive delivery systems for revascularization therapy and translational vascular tissue engineering.


Assuntos
Alginatos/farmacologia , Células Endoteliais/citologia , Heparina/farmacologia , Células-Tronco Pluripotentes Induzidas/citologia , Microesferas , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Aorta/efeitos dos fármacos , Aorta/fisiologia , Preparações de Ação Retardada/farmacologia , Liberação Controlada de Fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Cinética , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Ratos Sprague-Dawley
13.
Bioact Mater ; 6(7): 2198-2220, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33553810

RESUMO

The mechanical environment and anisotropic structure of the heart modulate cardiac function at the cellular, tissue and organ levels. During myocardial infarction (MI) and subsequent healing, however, this landscape changes significantly. In order to engineer cardiac biomaterials with the appropriate properties to enhance function after MI, the changes in the myocardium induced by MI must be clearly identified. In this review, we focus on the mechanical and structural properties of the healthy and infarcted myocardium in order to gain insight about the environment in which biomaterial-based cardiac therapies are expected to perform and the functional deficiencies caused by MI that the therapy must address. From this understanding, we discuss epicardial therapies for MI inspired by the mechanics and anisotropy of the heart focusing on passive devices, which feature a biomaterials approach, and active devices, which feature robotic and cellular components. Through this review, a detailed analysis is provided in order to inspire further development and translation of epicardial therapies for MI.

14.
Tissue Eng Part A ; 27(11-12): 703-713, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33430704

RESUMO

With this work, we design alginate-based hydrogels for therapeutically directing revascularization and repair processes in vivo. We immobilize pleiotrophin (PTN) in injectable hydrogel formulations as the target factor to stimulate proangiogenic responses in endothelial cells. The optimized heparin-alginate/chitosan hydrogels, produced by internal crosslinking with calcium carbonate, show good biocompatibility and injectability and allow controlling the release of immobilized proteins in the subcutaneous tissue over a period of 7 days. In vitro assays, performed with translational human induced pluripotent stem cell-derived endothelial cells, and the in vivo Matrigel plug assay are conducted to demonstrate the angiogenic effects of PTN on endothelial cells. Our results indicate that PTN stimulates endothelial cell morphogenesis in vitro and the migration of endothelial cells and macrophages as soon as 4 days after injections of the developed hydrogels, promoting the formation of structures similar to the healthy granulation tissue, which is an indicator of healing in ischemic wounds. These studies provide the rationale for further investigating this novel therapeutic for pursuing increased vascular density for efficient regeneration of ischemic tissues, by leveraging the host endothelial cell population to initiate angiogenic and reparative processes in vivo. Impact statement Localized, sustained, and controlled delivery of angiogenic factors is crucial for enabling the formation of novel vascular networks in ischemic tissues. This study describes the development of an injectable heparin-alginate/collagen hydrogel for controlling the in vivo release and bioactivity of pleiotrophin (PTN), a heparin-binding factor with significant angiogenic activity. We demonstrate that PTN promotes angiogenesis in an in vitro model of hypoxia and in preclinical subcutaneous models. These results advance our understanding of PTN function in guiding therapeutic angiogenesis and are critical to inform the development of novel translational strategies for ischemic tissue repair and regeneration.


Assuntos
Heparina , Células-Tronco Pluripotentes Induzidas , Alginatos/farmacologia , Proteínas de Transporte , Citocinas , Células Endoteliais , Heparina/farmacologia , Humanos , Hidrogéis/farmacologia , Morfogênese , Neovascularização Fisiológica
15.
Tissue Eng Part A ; 27(19-20): 1290-1304, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33472529

RESUMO

Engineered tissues designed for translational applications in regenerative medicine require vascular networks to deliver oxygen and nutrients rapidly to the implanted cells. A limiting factor of in vivo translation is the rapid and successful inosculation, or connection, of host and implanted vascular networks and subsequent perfusion of the implant. An approach gaining favor in vascular tissue engineering is to provide instructive cues from the engineered tissue to enhance host vascular penetration and connection with the implant. Here, we use a novel in vitro platform based on the aortic ring assay to evaluate the impact of patterned, endothelialized vessels or growth factor release from engineered constructs on preinosculative vascular cell outgrowth from surrogate host tissue in a controlled, defined environment, and introduce robust tools for evaluating vascular morphogenesis and chemotaxis. We demonstrate the creation of engineered vessels at the arteriole scale, which develop basement membrane, exhibit tight junctions, and actively sprout into the surrounding bulk hydrogel. Vessel-containing constructs are co-cultured adjacent to rodent aortic rings, and the resulting heterocellular outgrowth is quantified. Cells originating from the aortic ring migrate preferentially toward constructs containing engineered vessels with 1.5-fold faster outgrowth kinetics, 2.5-fold increased cellular density, and 1.6-fold greater network formation versus control (no endothelial cells and growth factor-reduced culture medium). Growth factor release from constructs with nonendothelialized channels and in reduced factor medium equivalently stimulates sustained vascular outgrowth distance, cellular density, and network formation, akin to engineered vessels in endothelial growth medium 2 (EGM-2) medium. In conclusion, we show that three-dimensional endothelialized patterned vessels or growth factor release stimulate a robust, host-derived vascular cell chemotactic response at early time points critical for instructive angiogenic cues. Further, we developed robust, unbiased tools to quantify metrics of vascular morphogenesis and preinosculative heterocellular outgrowth from rat aortic rings and demonstrated the utility of our complex, controlled environment, heterocellular in vitro platform. Impact statement Using a novel in vitro platform, we show that engineered constructs with patterned vessels or angiogenic growth factor release, two methods of instructing host revascularization responses, equivalently improve early host-derived vascular outgrowth. Our platform leverages the aortic ring assay in a tissue engineering context to study preinosculative vascular cell chemotaxis from surrogate host vascular cells in response to paracrine cues from co-cultured engineered tissues using robust, open-source quantification tools. Our accessible and flexible platform enables translationally focused studies in revascularization using implantable therapeutics containing prepatterned vessels with greater environmental control than in vivo studies to advance vascular tissue engineering.


Assuntos
Quimiotaxia , Células Endoteliais , Animais , Técnicas de Cocultura , Hidrogéis , Ratos , Engenharia Tecidual
16.
Stem Cells Int ; 2020: 9363809, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32724316

RESUMO

Cardiac tissue engineering using hiPSC-derived cardiomyocytes is a promising avenue for cardiovascular regeneration, pharmaceutical drug development, cardiotoxicity evaluation, and disease modeling. Limitations to these applications still exist due in part to the need for more robust structural support, organization, and electromechanical function of engineered cardiac tissues. It is well accepted that heterotypic cellular interactions impact the phenotype of cardiomyocytes. The current study evaluates the functional effects of coculturing adult human cardiac fibroblasts (hCFs) in 3D engineered tissues on excitation and contraction with the goal of recapitulating healthy, nonarrhythmogenic myocardium in vitro. A small population (5% of total cell number) of hCFs in tissues improves tissue formation, material properties, and contractile function. However, two perturbations to the hCF population create disease-like phenotypes in engineered cardiac tissues. First, increasing the percentage of hCFs to 15% resulted in tissues with increased ectopic activity and spontaneous excitation rate. Second, hCFs undergo myofibroblast activation in traditional two-dimensional culture, and this altered phenotype ablated the functional benefits of hCFs when incorporated into engineered cardiac tissues. Taken together, the results of this study demonstrate that human cardiac fibroblast number and activation state modulate electromechanical function of hiPSC-cardiomyocytes and that a low percentage of quiescent hCFs are a valuable cell source to advance a healthy electromechanical response of engineered cardiac tissue for regenerative medicine applications.

17.
Biomaterials ; 251: 120033, 2020 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-32388033

RESUMO

Heart regeneration after myocardial infarction requires new cardiomyocytes and a supportive vascular network. Here, we evaluate the efficacy of localized delivery of angiogenic factors from biomaterials within the implanted muscle tissue to guide growth of a more dense, organized, and perfused vascular supply into implanted engineered human cardiac tissue on an ischemia/reperfusion injured rat heart. We use large, aligned 3-dimensional engineered tissue with cardiomyocytes derived from human induced pluripotent stem cells in a collagen matrix that contains dispersed alginate microspheres as local protein depots. Release of angiogenic growth factors VEGF and bFGF in combination with morphogen sonic hedgehog from the microspheres into the local microenvironment occurs from the epicardial implant site. Analysis of the 3D vascular network in the engineered tissue via Microfil® perfusion and microCT imaging at 30 days shows increased volumetric network density with a wider distribution of vessel diameters, proportionally increased branching and length, and reduced tortuosity. Global heart function is increased in the angiogenic factor-loaded cardiac implants versus sham. These findings demonstrate for the first time the efficacy of a combined remuscularization and revascularization therapy for heart regeneration after myocardial infarction.

18.
Artigo em Inglês | MEDLINE | ID: mdl-32318563

RESUMO

Coronary artery disease is a severe ischemic condition characterized by the reduction of blood flow in the arteries of the heart that results in the dysfunction and death of cardiac tissue. Despite research over several decades on how to reduce long-term complications and promote angiogenesis in the infarct, the medical field has yet to define effective treatments for inducing revascularization in the ischemic tissue. With this work, we have developed functional biomaterials for the controlled release of immunomodulatory cytokines to direct immune cell fate for controlling wound healing in the ischemic myocardium. The reparative effects of colony-stimulating factor (CSF-1), and anti-inflammatory interleukins 4/6/13 (IL4/6/13) have been evaluated in vitro and in a predictive in vivo model of ischemia (the skin flap model) to optimize a new immunomodulatory biomaterial that we use for treating infarcted rat hearts. Alginate hydrogels have been produced by internal gelation with calcium carbonate (CaCO3) as carriers for the immunomodulatory cues, and their stability, degradation, rheological properties and release kinetics have been evaluated in vitro. CD14 positive human peripheral blood monocytes treated with the immunomodulatory biomaterials show polarization into pro-healing macrophage phenotypes. Unloaded and CSF-1/IL4 loaded alginate gel formulations have been implanted in skin flap ischemic wounds to test the safety and efficacy of the delivery system in vivo. Faster wound healing is observed with the new therapeutic treatment, compared to the wounds treated with the unloaded controls at day 14. The optimized therapy has been evaluated in a rat model of myocardial infarct (ischemia/reperfusion). Macrophage polarization toward healing phenotypes and global cardiac function measured with echocardiography and immunohistochemistry at 4 and 15 days demonstrate the therapeutic potential of the proposed immunomodulatory treatment in a clinically relevant infarct model.

19.
PLoS One ; 15(3): e0230001, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32155214

RESUMO

Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes are a valuable resource for cardiac therapeutic development; however, generation of these cells in large numbers and high purity is a limitation in widespread adoption. Here, design of experiments (DOE) is used to investigate the cardiac differentiation space of three hiPSC lines when varying CHIR99027 concentration and cell seeding density, and a novel image analysis is developed to evaluate plate coverage when initiating differentiation. Metabolic selection via lactate purifies hiPSC-cardiomyocyte populations, and the bioenergetic phenotype and engineered tissue mechanics of purified and unpurified hiPSC-cardiomyocytes are compared. Findings demonstrate that when initiating differentiation one day after hiPSC plating, low (3 µM) Chiron and 72 x 103 cells/cm2 seeding density result in peak cardiac purity (50-90%) for all three hiPSC lines. Our results confirm that metabolic selection with lactate shifts hiPSC-cardiomyocyte metabolism towards oxidative phosphorylation, but this more "mature" metabolic phenotype does not by itself result in a more mature contractile phenotype in engineered cardiac tissues at one week of culture in 3D tissues. This study provides widely adaptable methods including novel image analysis code and parameters for refining hiPSC-cardiomyocyte differentiation and describes the practical implications of metabolic selection of cardiomyocytes for downstream tissue engineering applications.


Assuntos
Diferenciação Celular , Técnicas Citológicas/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Adulto , Linhagem Celular , Humanos , Miócitos Cardíacos/metabolismo , Fenótipo
20.
J Biomed Mater Res B Appl Biomater ; 108(6): 2407-2416, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31984665

RESUMO

Engineering an angiogenic material for regenerative medicine requires knowledge of native extracellular matrix remodeling by cellular processes in angiogenesis. Vascularization remains a key challenge in the field of tissue engineering, one that can be mitigated by developing platforms conducive to guiding dynamic cell-matrix interactions required for new vessel formation. In this review, we highlight nuanced processes of angiogenesis and demonstrate how materials engineering is being used to interface with dynamic type I collagen remodeling, Notch and VEGF signaling, cell migration, and tissue morphogenesis. Because α1(I)-collagen is secreted by endothelial tip cells during sprouting angiogenesis and required for migration, collagen is a very useful natural biomaterial and its angiogenic modifications are described. The balance between collagen types I and IV via secretion and degradation is tightly controlled by proteinases and other cell types that are capable of internalizing collagen to maintain tissue integrity. Thus, we provide examples in skin and cardiac tissue engineering of collagen tailoring in diverse cellular microenvironments for tissue regeneration. As our understanding of how to drive collagen remodeling and cellular phenotype through angiogenic pathways grows, our capabilities to model and manipulate material systems must continue to expand to develop novel applications for wound healing, angiogenic therapy, and regenerative medicine.


Assuntos
Colágeno/química , Engenharia Metabólica/métodos , Neovascularização Fisiológica , Medicina Regenerativa/métodos , Animais , Humanos , Engenharia Tecidual , Alicerces Teciduais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...