Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(40): e2404644121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39312653

RESUMEN

With current plans for manned missions to Mars and beyond, the need to better understand, prevent, and counteract the harmful effects of long-duration spaceflight on the body is becoming increasingly important. In this study, an automated heart-on-a-chip platform was flown to the International Space Station on a 1-mo mission during which contractile cardiac function was monitored in real-time. Upon return to Earth, engineered human heart tissues (EHTs) were further analyzed with ultrastructural imaging and RNA sequencing to investigate the impact of prolonged microgravity on cardiomyocyte function and health. Spaceflight EHTs exhibited significantly reduced twitch forces, increased incidences of arrhythmias, and increased signs of sarcomere disruption and mitochondrial damage. Transcriptomic analyses showed an up-regulation of genes and pathways associated with metabolic disorders, heart failure, oxidative stress, and inflammation, while genes related to contractility and calcium signaling showed significant down-regulation. Finally, in silico modeling revealed a potential link between oxidative stress and mitochondrial dysfunction that corresponded with RNA sequencing results. This represents an in vitro model to faithfully reproduce the adverse effects of spaceflight on three-dimensional (3D)-engineered heart tissue.


Asunto(s)
Contracción Miocárdica , Miocitos Cardíacos , Vuelo Espacial , Vuelo Espacial/métodos , Humanos , Contracción Miocárdica/fisiología , Miocitos Cardíacos/metabolismo , Dispositivos Laboratorio en un Chip , Ingravidez/efectos adversos , Estrés Oxidativo , Mitocondrias/metabolismo , Mitocondrias Cardíacas/metabolismo
2.
Int J Mol Sci ; 25(17)2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39273136

RESUMEN

One of the many unresolved obstacles in the field of cardiovascular research is an uncompromising in vitro cardiac model. While primary cell sources from animal models offer both advantages and disadvantages, efforts over the past half-century have aimed to reduce their use. Additionally, obtaining a sufficient quantity of human primary cardiomyocytes faces ethical and legal challenges. As the practically unlimited source of human cardiomyocytes from induced pluripotent stem cells (hiPSC-CM) is now mostly resolved, there are great efforts to improve their quality and applicability by overcoming their intrinsic limitations. The greatest bottleneck in the field is the in vitro ageing of hiPSC-CMs to reach a maturity status that closely resembles that of the adult heart, thereby allowing for more appropriate drug developmental procedures as there is a clear correlation between ageing and developing cardiovascular diseases. Here, we review the current state-of-the-art techniques in the most realistic heart models used in disease modelling and toxicity evaluations from hiPSC-CM maturation through heart-on-a-chip platforms and in silico models to the in vitro models of certain cardiovascular diseases.


Asunto(s)
Cardiotoxicidad , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Cardiotoxicidad/etiología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/metabolismo , Animales , Diferenciación Celular , Enfermedades Cardiovasculares , Modelos Cardiovasculares
3.
Anal Bioanal Chem ; 415(18): 3911-3925, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36867198

RESUMEN

Cardiovascular diseases are a group of heart and blood vessel disorders which remain a leading cause of morbidity and mortality worldwide. Currently, cardiovascular disease research commonly depends on in vivo rodent models and in vitro human cell culture models. Despite their widespread use in cardiovascular disease research, there are some long-standing limitations: animal models often fail to faithfully mimic human response, while traditional cell models ignore the in vivo microenvironment, intercellular communications, and tissue-tissue interactions. The convergence of microfabrication and tissue engineering has given rise to organ-on-a-chip technologies. The organ-on-a-chip is a microdevice containing microfluidic chips, cells, and extracellular matrix to reproduce the physiological processes of a certain part of the human body, and is nowadays considered a promising bridge between in vivo models and in vitro 2D or 3D cell culture models. Considering the difficulty in obtaining human vessel and heart samples, the development of vessel-on-a-chip and heart-on-a-chip systems can guide cardiovascular disease research in the future. In this review, we elaborate methods and materials to fabricate organ-on-a-chip systems and summarize the construction of vessel and heart chips. The construction of vessels-on-a-chip must consider the cyclic mechanical stretch and fluid shear stress, while hemodynamic forces and cardiomyocyte maturation are key factors in building hearts-on-a-chip. We also introduce the application of organs-on-a-chip in cardiovascular disease study.


Asunto(s)
Enfermedades Cardiovasculares , Animales , Humanos , Sistemas Microfisiológicos , Microfluídica/métodos , Ingeniería de Tejidos/métodos , Tecnología , Dispositivos Laboratorio en un Chip
4.
Pflugers Arch ; 473(7): 1061-1085, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33629131

RESUMEN

Development of new drugs is of high interest for the field of cardiac and cardiovascular diseases, which are a dominant cause of death worldwide. Before being allowed to be used and distributed, every new potentially therapeutic compound must be strictly validated during preclinical and clinical trials. The preclinical studies usually involve the in vitro and in vivo evaluation. Due to the increasing reporting of discrepancy in drug effects in animal and humans and the requirement to reduce the number of animals used in research, improvement of in vitro models based on human cells is indispensable. Primary cardiac cells are difficult to access and maintain in cell culture for extensive experiments; therefore, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) became an excellent alternative. This technology enables a production of high number of patient- and disease-specific cardiomyocytes and other cardiac cell types for a large-scale research. The drug effects can be extensively evaluated in the context of electrophysiological responses with a use of well-established tools, such as multielectrode array (MEA), patch clamp, or calcium ion oscillation measurements. Cardiotoxicity, which is a common reason for withdrawing drugs from marketing or rejection at final stages of clinical trials, can be easily verified with a use of hiPSC-CM model providing a prediction of human-specific responses and higher safety of clinical trials involving patient cohort. Abovementioned studies can be performed using two-dimensional cell culture providing a high-throughput and relatively lower costs. On the other hand, more complex structures, such as engineered heart tissue, organoids, or spheroids, frequently applied as co-culture systems, represent more physiological conditions and higher maturation rate of hiPSC-derived cells. Furthermore, heart-on-a-chip technology has recently become an increasingly popular tool, as it implements controllable culture conditions, application of various stimulations and continuous parameters read-out. This paper is an overview of possible use of cardiomyocytes and other cardiac cell types derived from hiPSC as in vitro models of heart in drug research area prepared on the basis of latest scientific reports and providing thorough discussion regarding their advantages and limitations.


Asunto(s)
Corazón/fisiología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/citología , Animales , Desarrollo de Medicamentos/métodos , Humanos , Dispositivos Laboratorio en un Chip , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/fisiología
5.
Clin Sci (Lond) ; 131(13): 1393-1404, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28645929

RESUMEN

Engineering functional cardiac tissues remains an ongoing significant challenge due to the complexity of the native environment. However, our growing understanding of key parameters of the in vivo cardiac microenvironment and our ability to replicate those parameters in vitro are resulting in the development of increasingly sophisticated models of engineered cardiac tissues (ECT). This review examines some of the most relevant parameters that may be applied in culture leading to higher fidelity cardiac tissue models. These include the biochemical composition of culture media and cardiac lineage specification, co-culture conditions, electrical and mechanical stimulation, and the application of hydrogels, various biomaterials, and scaffolds. The review will also summarize some of the recent functional human tissue models that have been developed for in vivo and in vitro applications. Ultimately, the creation of sophisticated ECT that replicate native structure and function will be instrumental in advancing cell-based therapeutics and in providing advanced models for drug discovery and testing.


Asunto(s)
Miocardio/citología , Miocitos Cardíacos/citología , Ingeniería de Tejidos/métodos , Células Cultivadas , Técnicas de Cocultivo , Estimulación Eléctrica/métodos , Humanos , Hidrogeles , Modelos Cardiovasculares , Miocitos Cardíacos/fisiología , Estimulación Física/métodos , Andamios del Tejido
6.
J Biomed Mater Res A ; 112(4): 492-511, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-37909362

RESUMEN

Recent advances in both cardiac tissue engineering and hearts-on-a-chip are grounded in new biomaterial development as well as the employment of innovative fabrication techniques that enable precise control of the mechanical, electrical, and structural properties of the cardiac tissues being modelled. The elongated structure of cardiomyocytes requires tuning of substrate properties and application of biophysical stimuli to drive its mature phenotype. Landmark advances have already been achieved with induced pluripotent stem cell-derived cardiac patches that advanced to human testing. Heart-on-a-chip platforms are now commonly used by a number of pharmaceutical and biotechnology companies. Here, we provide an overview of cardiac physiology in order to better define the requirements for functional tissue recapitulation. We then discuss the biomaterials most commonly used in both cardiac tissue engineering and heart-on-a-chip, followed by the discussion of recent representative studies in both fields. We outline significant challenges common to both fields, specifically: scalable tissue fabrication and platform standardization, improving cellular fidelity through effective tissue vascularization, achieving adult tissue maturation, and ultimately developing cryopreservation protocols so that the tissues are available off the shelf.


Asunto(s)
Células Madre Pluripotentes Inducidas , Ingeniería de Tejidos , Humanos , Ingeniería de Tejidos/métodos , Miocitos Cardíacos , Materiales Biocompatibles , Dispositivos Laboratorio en un Chip , Miocardio
7.
Adv Healthc Mater ; 13(1): e2301338, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37471526

RESUMEN

Cardiovascular disease is a major cause of mortality worldwide, and current preclinical models including traditional animal models and 2D cell culture models have limitations in replicating human native heart physiology and response to drugs. Heart-on-a-chip (HoC) technology offers a promising solution by combining the advantages of cardiac tissue engineering and microfluidics to create in vitro 3D cardiac models, which can mimic key aspects of human microphysiological systems and provide controllable microenvironments. Herein, recent advances in HoC technologies are introduced, including engineered cardiac microtissue construction in vitro, microfluidic chip fabrication, microenvironmental stimulation, and real-time feedback systems. The development of cardiac tissue engineering methods is focused for 3D microtissue preparation, advanced strategies for HoC fabrication, and current applications of these platforms. Major challenges in HoC fabrication are discussed and the perspective on the potential for these platforms is provided to advance research and clinical applications.


Asunto(s)
Corazón , Ingeniería de Tejidos , Animales , Humanos , Corazón/fisiología , Microfluídica , Técnicas de Cultivo de Célula , Dispositivos Laboratorio en un Chip
8.
Bioact Mater ; 33: 46-60, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38024233

RESUMEN

The successful translation of organ-on-a-chip devices requires the development of an automated workflow for device fabrication, which is challenged by the need for precise deposition of multiple classes of materials in micro-meter scaled configurations. Many current heart-on-a-chip devices are produced manually, requiring the expertise and dexterity of skilled operators. Here, we devised an automated and scalable fabrication method to engineer a Biowire II multiwell platform to generate human iPSC-derived cardiac tissues. This high-throughput heart-on-a-chip platform incorporated fluorescent nanocomposite microwires as force sensors, produced from quantum dots and thermoplastic elastomer, and 3D printed on top of a polystyrene tissue culture base patterned by hot embossing. An array of built-in carbon electrodes was embedded in a single step into the base, flanking the microwells on both sides. The facile and rapid 3D printing approach efficiently and seamlessly scaled up the Biowire II system from an 8-well chip to a 24-well and a 96-well format, resulting in an increase of platform fabrication efficiency by 17,5000-69,000% per well. The device's compatibility with long-term electrical stimulation in each well facilitated the targeted generation of mature human iPSC-derived cardiac tissues, evident through a positive force-frequency relationship, post-rest potentiation, and well-aligned sarcomeric apparatus. This system's ease of use and its capacity to gauge drug responses in matured cardiac tissue make it a powerful and reliable platform for rapid preclinical drug screening and development.

9.
ACS Nano ; 18(1): 829-838, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38153966

RESUMEN

Cardiac fibrosis acts as a serious worldwide health issue due to its prevalence in numerous forms of cardiac disease and its essential link to cardiac failure. Considering the efficiency of stem cell therapy for cardiac fibrosis, great efforts have been dedicated to developing accurate models for investigating their underlying therapeutic mechanisms. Herein we present an elaborate biomimetic cardiac fibrosis-on-a-chip based on Janus structural color film (SCF) to provide microphysiological visuals for stem cell therapeutic studies. By coculturing cardiomyocytes (CMs) and cardiac fibroblasts (FBs) on Janus SCF with fibrosis induction, the chip can recreate physiological intercellular crosstalk within the fibrotic microenvironment, elucidating the physiological alterations of fibrotic hearts. In particular, the Janus structural color film possesses superior perceptual capabilities for capturing and responding to a weak cardiac force, demonstrating synchronized structural color shifts. Based on these features, we have not only explored the dynamic relationship between color mapping and the evaluated disease phenotype but also demonstrated the self-reporting capacity of the cardiac fibrosis-on-a-chip for the assessment of mesenchymal stem cell-derived exosome therapy. These features suggest that such a chip can potentially facilitate the evolution of precision medicine strategies and create a protocol for preclinical cardiac drug screening.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Humanos , Biomimética , Miocitos Cardíacos/patología , Fibrosis , Dispositivos Laboratorio en un Chip
10.
Polymers (Basel) ; 16(18)2024 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-39339128

RESUMEN

A major challenge in myocardial tissue engineering is replicating the heart's highly complex three-dimensional (3D) anisotropic structure. Heart-on-a-chip (HOC) is an emerging technology for constructing myocardial tissue in vitro in recent years, but most existing HOC systems face difficulties in constructing 3D myocardial tissue aligned with multiple cell layers. Electrospun nanofibers are commonly used as scaffolds for cell growth in myocardial tissue engineering, which can structurally simulate the extracellular matrix to induce the aligned growth of myocardial cells. Here, we developed an HOC that integrates multi-layered aligned polycaprolactone (PCL) nanofiber scaffolds inside microfluidic chips, and constructed 3D thick and aligned tissue with a layered seeding approach. By culturing human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) on chip, the myocardial tissue on the two layered nanofibers reached a thickness of ~53 µm compared with ~19 µm for single-layered nanofibers. The obtained myocardial tissue presented well-aligned structures, with densely distributed α-actinin. By the third day post seeding, the hiPSC-CMs contract highly synchronously, with a contraction frequency of 18 times/min. The HOC with multi-layered biomimetic scaffolds provided a dynamic in vitro culture environment for hiPSC-CMs. Together with the layered cell-seeding process, the designed HOC promoted the formation of thick, well-aligned myocardial tissue.

11.
Biofabrication ; 16(4)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39226913

RESUMEN

The fabrication of complex and stable vasculature in engineered cardiac tissues represents a significant hurdle towards building physiologically relevant models of the heart. Here, we implemented a 3D model of cardiac vasculogenesis, incorporating endothelial cells (EC), stromal cells, and human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) in a fibrin hydrogel. The presence of CMs disrupted vessel formation in 3D tissues, resulting in the upregulation of endothelial activation markers and altered extracellular vesicle (EV) signaling in engineered tissues as determined by the proteomic analysis of culture supernatant. miRNA sequencing of CM- and EC-secreted EVs highlighted key EV-miRNAs that were postulated to play differing roles in cardiac vasculogenesis, including the let-7 family and miR-126-3p in EC-EVs. In the absence of CMs, the supplementation of CM-EVs to EC monolayers attenuated EC migration and proliferation and resulted in shorter and more discontinuous self-assembling vessels when applied to 3D vascular tissues. In contrast, supplementation of EC-EVs to the tissue culture media of 3D vascularized cardiac tissues mitigated some of the deleterious effects of CMs on vascular self-assembly, enhancing the average length and continuity of vessel tubes that formed in the presence of CMs. Direct transfection validated the effects of the key EC-EV miRNAs let-7b-5p and miR-126-3p in improving the maintenance of continuous vascular networks. EC-EV supplementation to biofabricated cardiac tissues and microfluidic devices resulted in tissue vascularization, illustrating the use of this approach in the engineering of enhanced, perfusable, microfluidic models of the myocardium.


Asunto(s)
Vesículas Extracelulares , Células Madre Pluripotentes Inducidas , MicroARNs , Miocitos Cardíacos , Ingeniería de Tejidos , Humanos , Vesículas Extracelulares/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , MicroARNs/metabolismo , MicroARNs/genética , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/citología , Neovascularización Fisiológica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proliferación Celular , Miocardio/metabolismo , Miocardio/citología
12.
Adv Healthc Mater ; 13(21): e2302642, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38683053

RESUMEN

Epicardial cells (EPIs) form the outer layer of the heart and play an important role in development and disease. Current heart-on-a-chip platforms still do not fully mimic the native cardiac environment due to the absence of relevant cell types, such as EPIs. Here, using the Biowire II platform, engineered cardiac tissues with an epicardial outer layer and inner myocardial structure are constructed, and an image analysis approach is developed to track the EPI cell migration in a beating myocardial environment. Functional properties of EPI cardiac tissues improve over two weeks in culture. In conditions mimicking ischemia reperfusion injury (IRI), the EPI cardiac tissues experience less cell death and a lower impact on functional properties. EPI cell coverage is significantly reduced and more diffuse under normoxic conditions compared to the post-IRI conditions. Upon IRI, migration of EPI cells into the cardiac tissue interior is observed, with contributions to alpha smooth muscle actin positive cell population. Altogether, a novel heart-on-a-chip model is designed to incorporate EPIs through a formation process that mimics cardiac development, and this work demonstrates that EPI cardiac tissues respond to injury differently than epicardium-free controls, highlighting the importance of including EPIs in heart-on-a-chip constructs that aim to accurately mimic the cardiac environment.


Asunto(s)
Dispositivos Laboratorio en un Chip , Pericardio , Pericardio/metabolismo , Animales , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Movimiento Celular , Miocardio/metabolismo , Miocardio/patología , Ingeniería de Tejidos/métodos , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
13.
Mater Today Bio ; 24: 100914, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38179431

RESUMEN

Recent advances in heart-on-a-chip systems hold great promise to facilitate cardiac physiological, pathological, and pharmacological studies. This review focuses on the development of heart-on-a-chip systems with tissue-specific functionalities. For one thing, the strategies for developing cardiac microtissues on heart-on-a-chip systems that closely mimic the structures and behaviors of the native heart are analyzed, including the imitation of cardiac structural and functional characteristics. For another, the development of techniques for real-time monitoring of biophysical and biochemical signals from cardiac microtissues on heart-on-a-chip systems is introduced, incorporating cardiac electrophysiological signals, contractile activity, and biomarkers. Furthermore, the applications of heart-on-a-chip systems in intelligent cardiac studies are discussed regarding physiological/pathological research and pharmacological assessment. Finally, the future development of heart-on-a-chip toward a higher level of systematization, integration, and maturation is proposed.

14.
Bioengineering (Basel) ; 11(3)2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38534508

RESUMEN

The implementation of three-dimensional tissue engineering concurrently with stem cell technology holds great promise for in vitro research in pharmacology and toxicology and modeling cardiac diseases, particularly for rare genetic and pediatric diseases for which animal models, immortal cell lines, and biopsy samples are unavailable. It also allows for a rapid assessment of phenotype-genotype relationships and tissue response to pharmacological manipulation. Mutations in the TSC1 and TSC2 genes lead to dysfunctional mTOR signaling and cause tuberous sclerosis complex (TSC), a genetic disorder that affects multiple organ systems, principally the brain, heart, skin, and kidneys. Here we differentiated healthy (CC3) and tuberous sclerosis (TSP8-15) human induced pluripotent stem cells (hiPSCs) into cardiomyocytes to create engineered cardiac tissue constructs (ECTCs). We investigated and compared their mechano-elastic properties and gene expression and assessed the effects of rapamycin, a potent inhibitor of the mechanistic target of rapamycin (mTOR). The TSP8-15 ECTCs had increased chronotropy compared to healthy ECTCs. Rapamycin induced positive inotropic and chronotropic effects (i.e., increased contractility and beating frequency, respectively) in the CC3 ECTCs but did not cause significant changes in the TSP8-15 ECTCs. A differential gene expression analysis revealed 926 up- and 439 down-regulated genes in the TSP8-15 ECTCs compared to their healthy counterparts. The application of rapamycin initiated the differential expression of 101 and 31 genes in the CC3 and TSP8-15 ECTCs, respectively. A gene ontology analysis showed that in the CC3 ECTCs, the positive inotropic and chronotropic effects of rapamycin correlated with positively regulated biological processes, which were primarily related to the metabolism of lipids and fatty and amino acids, and with negatively regulated processes, which were predominantly associated with cell proliferation and muscle and tissue development. In conclusion, this study describes for the first time an in vitro TSC cardiac tissue model, illustrates the response of normal and TSC ECTCs to rapamycin, and provides new insights into the mechanisms of TSC.

15.
Sci Rep ; 14(1): 18063, 2024 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117679

RESUMEN

In recent years, research on organ-on-a-chip technology has been flourishing, particularly for drug screening and disease model development. Fibroblasts and vascular endothelial cells engage in crosstalk through paracrine signaling and direct cell-cell contact, which is essential for the normal development and function of the heart. Therefore, to faithfully recapitulate cardiac function, it is imperative to incorporate fibroblasts and vascular endothelial cells into a heart-on-a-chip model. Here, we report the development of a human heart-on-a-chip composed of induced pluripotent stem cell (iPSC)-derived cardiomyocytes, fibroblasts, and vascular endothelial cells. Vascular endothelial cells cultured on microfluidic channels responded to the flow of culture medium mimicking blood flow by orienting themselves parallel to the flow direction, akin to in vivo vascular alignment in response to blood flow. Furthermore, the flow of culture medium promoted integrity among vascular endothelial cells, as evidenced by CD31 staining and lower apparent permeability. The tri-culture condition of iPSC-derived cardiomyocytes, fibroblasts, and vascular endothelial cells resulted in higher expression of the ventricular cardiomyocyte marker IRX4 and increased contractility compared to the bi-culture condition with iPSC-derived cardiomyocytes and fibroblasts alone. Such tri-culture-derived cardiac tissues exhibited cardiac responses similar to in vivo hearts, including an increase in heart rate upon noradrenaline administration. In summary, we have achieved the development of a heart-on-a-chip composed of cardiomyocytes, fibroblasts, and vascular endothelial cells that mimics in vivo cardiac behavior.


Asunto(s)
Células Endoteliales , Fibroblastos , Células Madre Pluripotentes Inducidas , Dispositivos Laboratorio en un Chip , Miocitos Cardíacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Fibroblastos/citología , Fibroblastos/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo/métodos , Sistemas Microfisiológicos
16.
Cells ; 13(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38334642

RESUMEN

The human heart lacks significant regenerative capacity; thus, the solution to heart failure (HF) remains organ donation, requiring surgery and immunosuppression. The demand for constructed cardiac tissues (CCTs) to model and treat disease continues to grow. Recent advances in induced pluripotent stem cell (iPSC) manipulation, CRISPR gene editing, and 3D tissue culture have enabled a boom in iPSC-derived CCTs (iPSC-CCTs) with diverse cell types and architecture. Compared with 2D-cultured cells, iPSC-CCTs better recapitulate heart biology, demonstrating the potential to advance organ modeling, drug discovery, and regenerative medicine, though iPSC-CCTs could benefit from better methods to faithfully mimic heart physiology and electrophysiology. Here, we summarize advances in iPSC-CCTs and future developments in the vascularization, immunization, and maturation of iPSC-CCTs for study and therapy.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Corazón/fisiología , Medicina Regenerativa , Descubrimiento de Drogas
17.
ACS Nano ; 17(15): 15180-15188, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37459507

RESUMEN

Environmental toxins can result in serious and fatal damage in the human heart, while the development of a viable stratagem for assessing the effects of environmental toxins on human cardiac tissue is still a challenge. Herein, we present a heart-on-a-chip based on human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) cultured living anisotropic structural color hydrogels for cardiotoxicity screening. Such anisotropic structural color hydrogels with a conductive parallel carbon nanotube (CNT) upper layer, gelatin methacryloyl (GelMA) interlayer, and inverse opal bottom layer were fabricated by a sandwich replicating approach. The inverse opal structure endowed the anisotropic hydrogels with stable structural color property, while the parallel and conductive CNTs could induce the hiPSC-CMs to grow in a directional manner with consistent autonomous beating. Notably, the resultant hiPSC-CM-cultured hydrogel exhibited synchronous shifts in structural color, responding to contraction and relaxation of hiPSC-CMs, offering a visual platform for monitoring cell activity. Given these features, the hiPSC-CM-cultured living anisotropic structural color hydrogels were integrated into a heart-on-a-chip, which provided a superior cardiotoxicity screening platform for environmental toxins.


Asunto(s)
Células Madre Pluripotentes Inducidas , Nanotubos de Carbono , Humanos , Cardiotoxicidad , Hidrogeles/química , Miocitos Cardíacos , Nanotubos de Carbono/toxicidad , Células Cultivadas
18.
Biosens Bioelectron ; 220: 114840, 2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-36402101

RESUMEN

Heart disease is the leading cause of death worldwide and imposes a significant burden on healthcare systems globally. A major hurdle to the development of more effective therapeutics is the reliance on animal models that fail to faithfully recapitulate human pathophysiology. The predictivity of in vitro models that lack the complexity of in vivo tissue remain poor as well. To combat these issues, researchers are developing organ-on-a-chip models of the heart that leverage the use of human induced pluripotent stem cell-derived cardiomyocytes in combination with novel platforms engineered to better recapitulate tissue- and organ-level physiology. The integration of novel biosensors into these platforms is also a critical step in the development of these models, as they allow for increased throughput, real-time and longitudinal phenotypic assessment, and improved efficiency during preclinical disease modeling and drug screening studies. These platforms hold great promise for both improving our understanding of heart disease as well as for screening potential therapeutics based on clinically relevant endpoints with better predictivity of clinical outcomes. In this review, we describe state-of-the-art heart-on-a-chip platforms, the integration of novel biosensors into these models for real-time and continual monitoring of tissue-level physiology, as well as their use for modeling heart disease and drug screening applications. We also discuss future perspectives and further advances required to enable clinical trials-on-a-chip and next-generation precision medicine platforms.


Asunto(s)
Técnicas Biosensibles , Cardiopatías , Células Madre Pluripotentes Inducidas , Animales , Humanos , Evaluación Preclínica de Medicamentos , Dispositivos Laboratorio en un Chip , Cardiopatías/diagnóstico , Cardiopatías/tratamiento farmacológico , Miocitos Cardíacos
19.
Sci Bull (Beijing) ; 68(9): 938-945, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37062651

RESUMEN

Heart-on-chips have emerged as a powerful tool to promote the paradigm innovation in cardiac pathological research and drug development. Attempts are focused on improving microphysiological visuals, enhancing bionic characteristics, as well as expanding their biomedical applications. Herein, inspired by the bright feathers of peacock, we present a novel optical and electrical dual-responsive heart-on-a-chip based on cardiomyocytes hybrid bright MXene structural color hydrogels for hormone toxicity evaluation. Such hydrogels with inverse opal nanostructure are generated by using pregel to replicate MXene-decorated colloidal photonic crystal (PhC) array templates. The attendant MXene in the hydrogels could not only enhance the saturation of structural color, but also ensure the composite hydrogel with excellent electroconductivity to facilitate the synergetic beating of their surface cultured cardiomyocytes. In this case, the hydrogels would undergo a synchronous deformation and generate shift in corresponding photonic band gap and structural color, which could be employed as visual signal for self-reporting of the cardiomyocyte mechanics. Based on these features, we demonstrated the practical value of the optical and electrical dual-responsive structural color MXene hydrogels constructed heart-on-a-chip in hormone toxicity testing. These results indicated that the proposed heart-on-a-chip might find broad prospects in drug screening, biological research, and so on.


Asunto(s)
Hidrogeles , Nanoestructuras , Animales , Hidrogeles/química , Miocitos Cardíacos , Nanoestructuras/uso terapéutico , Dispositivos Laboratorio en un Chip
20.
Biofabrication ; 15(4)2023 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-37267929

RESUMEN

Cardiovascular diseases (CVDs) are a major cause of death worldwide, leading to increased medical care costs. To turn the scale, it is essential to acquire a more in-depth and comprehensive understanding of CVDs and thus formulate more efficient and reliable treatments. Over the last decade, tremendous effort has been made to develop microfluidic systems to recapitulate native cardiovascular environments because of their unique advantages over conventional 2D culture systems and animal models such as high reproductivity, physiological relevance, and good controllability. These novel microfluidic systems could be extensively adopted for natural organ simulation, disease modeling, drug screening, disease diagnosis and therapy. Here, a brief review of the innovative designs of microfluidic devices for CVDs research is presented, with specific discussions on material selection, critical physiological and physical considerations. In addition, we elaborate on various biomedical applications of these microfluidic systems such as blood-vessel-on-a-chip and heart-on-a-chip, which are conducive to the investigation of the underlying mechanisms of CVDs. This review also provides systematic guidance on the construction of next-generation microfluidic systems for the diagnosis and treatment of CVDs. Finally, the challenges and future directions in this field are highlighted and discussed.


Asunto(s)
Enfermedades Cardiovasculares , Animales , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/terapia , Sistemas Microfisiológicos , Microfluídica , Dispositivos Laboratorio en un Chip , Corazón
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda