Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biofabrication ; 16(4)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38906132

RESUMO

The ability to controllably perfuse kidney organoids would better recapitulate the native tissue microenvironment for applications ranging from drug testing to therapeutic use. Here, we report a perfusable, vascularized kidney organoid on chip model composed of two individually addressable channels embedded in an extracellular matrix (ECM). The channels are respectively seeded with kidney organoids and human umbilical vein endothelial cells that form a confluent endothelium (macrovessel). During perfusion, endogenous endothelial cells present within the kidney organoids migrate through the ECM towards the macrovessel, where they form lumen-on-lumen anastomoses that are supported by stromal-like cells. Once micro-macrovessel integration is achieved, we introduced fluorescently labeled dextran of varying molecular weight and red blood cells into the macrovessel, which are transported through the microvascular network to the glomerular epithelia within the kidney organoids. Our approach for achieving controlled organoid perfusion opens new avenues for generating other perfused human tissues.


Assuntos
Células Endoteliais da Veia Umbilical Humana , Rim , Organoides , Perfusão , Organoides/citologia , Humanos , Rim/citologia , Rim/irrigação sanguínea , Dispositivos Lab-On-A-Chip , Animais , Engenharia Tecidual/métodos , Matriz Extracelular/metabolismo
2.
Biofabrication ; 16(1)2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37734324

RESUMO

Rete ridges consist of undulations between the epidermis and dermis that enhance the mechanical properties and biological function of human skin. However, most human skin models are fabricated with a flat interface between the epidermal and dermal layers. Here, we report a micro-stamping method for producing human skin models patterned with rete ridges of controlled geometry. To mitigate keratinocyte-induced matrix degradation, telocollagen-fibrin matrices with and without crosslinks enable these micropatterned features to persist during longitudinal culture. Our human skin model exhibits an epidermis that includes the following markers: cytokeratin 14, p63, and Ki67 in the basal layer, cytokeratin 10 in the suprabasal layer, and laminin and collagen IV in the basement membrane. We demonstrated that two keratinocyte cell lines, one from a neonatal donor and another from an adult diabetic donor, are compatible with this model. We tested this model using an irritation test and showed that the epidermis prevents rapid penetration of sodium dodecyl sulfate. Gene expression analysis revealed differences in keratinocytes obtained from the two donors as well as between 2D (control) and 3D culture conditions. Our human skin model may find potential application for drug and cosmetic testing, disease and wound healing modeling, and aging studies.


Assuntos
Biomimética , Pele , Adulto , Recém-Nascido , Humanos , Epiderme , Queratinócitos , Derme
4.
Adv Mater ; 35(5): e2206958, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36404106

RESUMO

Recent advances in computational design and 3D printing enable the fabrication of polymer lattices with high strength-to-weight ratio and tailored mechanics. To date, 3D lattices composed of monolithic materials have primarily been constructed due to limitations associated with most commercial 3D printing platforms. Here, freeform fabrication of multi-material polymer lattices via embedded three-dimensional (EMB3D) printing is demonstrated. An algorithm is developed first that generates print paths for each target lattice based on graph theory. The effects of ink rheology on filamentary printing and the effects of the print path on resultant mechanical properties are then investigated. By co-printing multiple materials with different mechanical properties, a broad range of periodic and stochastic lattices with tailored mechanical responses can be realized opening new avenues for constructing architected matter.

5.
Transl Res ; 250: 1-17, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35750295

RESUMO

The kidney is a vital organ that regulates the bodily fluid and electrolyte homeostasis via tailored urinary excretion. Kidney injuries that cause severe or progressive chronic kidney disease have driven the growing population of patients with end-stage kidney disease, leading to substantial patient morbidity and mortality. This irreversible kidney damage has also created a huge socioeconomical burden on the healthcare system, highlighting the need for novel translational research models for progressive kidney diseases. Conventional research methods such as in vitro 2D cell culture or animal models do not fully recapitulate complex human kidney diseases. By contrast, directed differentiation of human induced pluripotent stem cells enables in vitro generation of patient-specific 3D kidney organoids, which can be used to model acute or chronic forms of hereditary, developmental, and metabolic kidney diseases. Furthermore, when combined with biofabrication techniques, organoids can be used as building blocks to construct vascularized kidney tissues mimicking their in vivo counterpart. By applying gene editing technology, organoid building blocks may be modified to minimize the process of immune rejection in kidney transplant recipients. In the foreseeable future, the universal kidney organoids derived from HLA-edited/deleted induced pluripotent stem cell (iPSC) lines may enable the supply of bioengineered organotypic kidney structures that are immune-compatible for the majority of the world population. Here, we summarize recent advances in kidney organoid research coupled with novel technologies such as organoids-on-chip and biofabrication of 3D kidney tissues providing convenient platforms for high-throughput drug screening, disease modelling, and therapeutic applications.


Assuntos
Células-Tronco Pluripotentes Induzidas , Insuficiência Renal Crônica , Animais , Humanos , Organoides , Rim , Diferenciação Celular , Insuficiência Renal Crônica/metabolismo
6.
Cell Stem Cell ; 29(5): 667-677, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523137

RESUMO

The construction of human organs on demand remains a tantalizing vision to solve the organ donor shortage. Yet, engineering tissues that recapitulate the cellular and architectural complexity of native organs is a grand challenge. The use of organ building blocks (OBBs) composed of multicellular spheroids, organoids, and assembloids offers an important pathway for creating organ-specific tissues with the desired cellular-to-tissue-level organization. Here, we review the differentiation, maturation, and 3D assembly of OBBs into functional human tissues and, ultimately, organs for therapeutic repair and replacement. We also highlight future challenges and areas of opportunity for this nascent field.


Assuntos
Organoides , Engenharia Tecidual , Humanos , Esferoides Celulares
7.
Adv Mater ; 34(26): e2200217, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35451188

RESUMO

The ability to replicate the 3D myocardial architecture found in human hearts is a grand challenge. Here, the fabrication of aligned cardiac tissues via bioprinting anisotropic organ building blocks (aOBBs) composed of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) is reported. A bioink composed of contractile cardiac aOBBs is first generated and aligned cardiac tissue sheets with linear, spiral, and chevron features are printed. Next, aligned cardiac macrofilaments are printed, whose contractile force and conduction velocity increase over time and exceed the performance of spheroid-based cardiac tissues. Finally, the ability to spatially control the magnitude and direction of contractile force by printing cardiac sheets with different aOBB alignment is highlighted. This research opens new avenues to generating functional cardiac tissue with high cell density and complex cellular alignment.


Assuntos
Bioimpressão , Células-Tronco Pluripotentes Induzidas , Humanos , Miocárdio , Miócitos Cardíacos , Impressão Tridimensional , Engenharia Tecidual , Alicerces Teciduais
8.
Nat Protoc ; 15(2): 421-449, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31932771

RESUMO

This protocol describes the design, fabrication and use of a 3D physiological and pathophysiological motor unit model consisting of motor neurons coupled to skeletal muscles interacting via the neuromuscular junction (NMJ) within a microfluidic device. This model facilitates imaging and quantitative functional assessment. The 'NMJ chip' enables real-time, live imaging of axonal outgrowth, NMJ formation and muscle maturation, as well as synchronization of motor neuron activity and muscle contraction under optogenetic control for the study of normal physiological events. The proposed protocol takes ~2-3 months to be implemented. Pathological behaviors associated with various neuromuscular diseases, such as regression of motor neuron axons, motor neuron death, and muscle degradation and atrophy can also be recapitulated in this system. Disease models can be created by the use of patient-derived induced pluripotent stem cells to generate both the motor neurons and skeletal muscle cells used. This is demonstrated by the use of cells from a patient with sporadic amyotrophic lateral sclerosis but can be applied more generally to models of neuromuscular disease, such as spinal muscular atrophy, NMJ disorder and muscular dystrophy. Models such as this hold considerable potential for applications in precision medicine, drug screening and disease risk assessment.


Assuntos
Avaliação Pré-Clínica de Medicamentos/instrumentação , Procedimentos Analíticos em Microchip/métodos , Doenças Neuromusculares/tratamento farmacológico , Medicina de Precisão/instrumentação , Humanos , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Doenças Neuromusculares/patologia , Doenças Neuromusculares/fisiopatologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Medição de Risco
9.
Sci Adv ; 5(9): eaaw2459, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31523707

RESUMO

Engineering organ-specific tissues for therapeutic applications is a grand challenge, requiring the fabrication and maintenance of densely cellular constructs composed of ~108 cells/ml. Organ building blocks (OBBs) composed of patient-specific-induced pluripotent stem cell-derived organoids offer a pathway to achieving tissues with the requisite cellular density, microarchitecture, and function. However, to date, scant attention has been devoted to their assembly into 3D tissue constructs. Here, we report a biomanufacturing method for assembling hundreds of thousands of these OBBs into living matrices with high cellular density into which perfusable vascular channels are introduced via embedded three-dimensional bioprinting. The OBB matrices exhibit the desired self-healing, viscoplastic behavior required for sacrificial writing into functional tissue (SWIFT). As an exemplar, we created a perfusable cardiac tissue that fuses and beats synchronously over a 7-day period. Our SWIFT biomanufacturing method enables the rapid assembly of perfusable patient- and organ-specific tissues at therapeutic scales.


Assuntos
Bioimpressão , Vasos Coronários/metabolismo , Matriz Extracelular/química , Células-Tronco Pluripotentes Induzidas/metabolismo , Miocárdio/metabolismo , Engenharia Tecidual , Vasos Coronários/citologia , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Miocárdio/citologia
10.
Theranostics ; 9(5): 1232-1246, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30867827

RESUMO

The study of human neuromuscular diseases has traditionally been performed in animal models, due to the difficulty of performing studies in human subjects. Despite the unquestioned value of animal models, inter-species differences hamper the translation of these findings to clinical trials. Tissue-engineered models of the neuromuscular junction (NMJ) allow for the recapitulation of the human physiology in tightly controlled in vitro settings. Methods: Here we report the first human patient-specific tissue-engineered model of the neuromuscular junction (NMJ) that combines stem cell technology with tissue engineering, optogenetics, microfabrication and image processing. The combination of custom-made hardware and software allows for repeated, quantitative measurements of NMJ function in a user-independent manner. Results: We demonstrate the utility of this model for basic and translational research by characterizing in real time the functional changes during physiological and pathological processes. Principal Conclusions: This system holds great potential for the study of neuromuscular diseases and drug screening, allowing for the extraction of quantitative functional data from a human, patient-specific system.


Assuntos
Modelos Teóricos , Doenças Neuromusculares/patologia , Doenças Neuromusculares/fisiopatologia , Optogenética/métodos , Engenharia Tecidual/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Junção Neuromuscular/patologia , Junção Neuromuscular/fisiologia , Junção Neuromuscular/fisiopatologia
11.
Sci Adv ; 4(10): eaat5847, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30324134

RESUMO

Amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disease involving loss of motor neurons (MNs) and muscle atrophy, still has no effective treatment, despite much research effort. To provide a platform for testing drug candidates and investigating the pathogenesis of ALS, we developed an ALS-on-a-chip technology (i.e., an ALS motor unit) using three-dimensional skeletal muscle bundles along with induced pluripotent stem cell (iPSC)-derived and light-sensitive channelrhodopsin-2-induced MN spheroids from a patient with sporadic ALS. Each tissue was cultured in a different compartment of a microfluidic device. Axon outgrowth formed neuromuscular junctions on the muscle fiber bundles. Light was used to activate muscle contraction, which was measured on the basis of pillar deflections. Compared to a non-ALS motor unit, the ALS motor unit generated fewer muscle contractions, there was MN degradation, and apoptosis increased in the muscle. Furthermore, the muscle contractions were recovered by single treatments and cotreatment with rapamycin (a mechanistic target of rapamycin inhibitor) and bosutinib (an Src/c-Abl inhibitor). This recovery was associated with up-regulation of autophagy and degradation of TAR DNA binding protein-43 in the MNs. Moreover, administering the drugs via an endothelial cell barrier decreased the expression of P-glycoprotein (an efflux pump that transports bosutinib) in the endothelial cells, indicating that rapamycin and bosutinib cotreatment has considerable potential for ALS treatment. This ALS-on-a-chip and optogenetics technology could help to elucidate the pathogenesis of ALS and to screen for drug candidates.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios Motores/patologia , Músculo Esquelético/citologia , Animais , Sinalização do Cálcio , Estimulação Elétrica , Expressão Gênica , Ácido Glutâmico/metabolismo , Ácido Glutâmico/farmacologia , Humanos , Dispositivos Lab-On-A-Chip , Camundongos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/fisiologia , Contração Muscular , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/fisiopatologia , Junção Neuromuscular/citologia , Junção Neuromuscular/fisiologia , Optogenética , Esferoides Celulares
12.
Adv Mater ; 30(15): e1706383, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29484726

RESUMO

Humans possess manual dexterity, motor skills, and other physical abilities that rely on feedback provided by the somatosensory system. Herein, a method is reported for creating soft somatosensitive actuators (SSAs) via embedded 3D printing, which are innervated with multiple conductive features that simultaneously enable haptic, proprioceptive, and thermoceptive sensing. This novel manufacturing approach enables the seamless integration of multiple ionically conductive and fluidic features within elastomeric matrices to produce SSAs with the desired bioinspired sensing and actuation capabilities. Each printed sensor is composed of an ionically conductive gel that exhibits both long-term stability and hysteresis-free performance. As an exemplar, multiple SSAs are combined into a soft robotic gripper that provides proprioceptive and haptic feedback via embedded curvature, inflation, and contact sensors, including deep and fine touch contact sensors. The multimaterial manufacturing platform enables complex sensing motifs to be easily integrated into soft actuating systems, which is a necessary step toward closed-loop feedback control of soft robots, machines, and haptic devices.

13.
Sci Adv ; 2(8): e1501429, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27493991

RESUMO

Motor units are the fundamental elements responsible for muscle movement. They are formed by lower motor neurons and their muscle targets, synapsed via neuromuscular junctions (NMJs). The loss of NMJs in neurodegenerative disorders (such as amyotrophic lateral sclerosis or spinal muscle atrophy) or as a result of traumatic injuries affects millions of lives each year. Developing in vitro assays that closely recapitulate the physiology of neuromuscular tissues is crucial to understand the formation and maturation of NMJs, as well as to help unravel the mechanisms leading to their degeneration and repair. We present a microfluidic platform designed to coculture myoblast-derived muscle strips and motor neurons differentiated from mouse embryonic stem cells (ESCs) within a three-dimensional (3D) hydrogel. The device geometry mimics the spinal cord-limb physical separation by compartmentalizing the two cell types, which also facilitates the observation of 3D neurite outgrowth and remote muscle innervation. Moreover, the use of compliant pillars as anchors for muscle strips provides a quantitative functional readout of force generation. Finally, photosensitizing the ESC provides a pool of source cells that can be differentiated into optically excitable motor neurons, allowing for spatiodynamic, versatile, and noninvasive in vitro control of the motor units.


Assuntos
Dispositivos Lab-On-A-Chip , Neurônios Motores/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Junção Neuromuscular/fisiologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Channelrhodopsins , Técnicas de Cocultura , Expressão Gênica , Genes Reporter , Técnicas In Vitro , Camundongos , Neurônios Motores/citologia , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Contração Muscular/genética , Fibras Musculares Esqueléticas/citologia , Engenharia Tecidual
14.
Proc Natl Acad Sci U S A ; 113(13): 3497-502, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26976577

RESUMO

Complex biological systems sense, process, and respond to their surroundings in real time. The ability of such systems to adapt their behavioral response to suit a range of dynamic environmental signals motivates the use of biological materials for other engineering applications. As a step toward forward engineering biological machines (bio-bots) capable of nonnatural functional behaviors, we created a modular light-controlled skeletal muscle-powered bioactuator that can generate up to 300 µN (0.56 kPa) of active tension force in response to a noninvasive optical stimulus. When coupled to a 3D printed flexible bio-bot skeleton, these actuators drive directional locomotion (310 µm/s or 1.3 body lengths/min) and 2D rotational steering (2°/s) in a precisely targeted and controllable manner. The muscle actuators dynamically adapt to their surroundings by adjusting performance in response to "exercise" training stimuli. This demonstration sets the stage for developing multicellular bio-integrated machines and systems for a range of applications.


Assuntos
Músculo Esquelético/fisiologia , Optogenética/métodos , Animais , Linhagem Celular , Desenho de Equipamento , Análise de Elementos Finitos , Locomoção , Camundongos , Contração Muscular/fisiologia , Optogenética/instrumentação , Impressão Tridimensional , Robótica/instrumentação , Robótica/métodos , Imagem com Lapso de Tempo , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos
15.
J Physiol ; 594(12): 3245-70, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-26864374

RESUMO

KEY POINTS: Xenopus laevis craniofacial development is a good system for the study of Andersen-Tawil Syndrome (ATS)-associated craniofacial anomalies (CFAs) because (1) Kcnj2 is expressed in the nascent face; (2) molecular-genetic and biophysical techniques are available for the study of ion-dependent signalling during craniofacial morphogenesis; (3) as in humans, expression of variant Kcnj2 forms in embryos causes a muscle phenotype; and (4) variant forms of Kcnj2 found in human patients, when injected into frog embryos, cause CFAs in the same cell lineages. Forced expression of WT or variant Kcnj2 changes the normal pattern of Vmem (resting potential) regionalization found in the ectoderm of neurulating embryos, and changes the normal pattern of expression of ten different genetic regulators of craniofacial development, including markers of cranial neural crest and of placodes. Expression of other potassium channels and two different light-activated channels, all of which have an effect on Vmem , causes CFAs like those induced by injection of Kcnj2 variants. In contrast, expression of Slc9A (NHE3), an electroneutral ion channel, and of GlyR, an inactive Cl(-) channel, do not cause CFAs, demonstrating that correct craniofacial development depends on a pattern of bioelectric states, not on ion- or channel-specific signalling. Using optogenetics to control both the location and the timing of ion flux in developing embryos, we show that affecting Vmem of the ectoderm and no other cell layers is sufficient to cause CFAs, but only during early neurula stages. Changes in Vmem induced late in neurulation do not affect craniofacial development. We interpret these data as strong evidence, consistent with our hypothesis, that ATS-associated CFAs are caused by the effect of variant Kcnj2 on the Vmem of ectodermal cells of the developing face. We predict that the critical time is early during neurulation, and the critical cells are the ectodermal cranial neural crest and placode lineages. This points to the potential utility of extant, ion flux-modifying drugs as treatments to prevent CFAs associated with channelopathies such as ATS. ABSTRACT: Variants in potassium channel KCNJ2 cause Andersen-Tawil Syndrome (ATS); the induced craniofacial anomalies (CFAs) are entirely unexplained. We show that KCNJ2 is expressed in Xenopus and mouse during the earliest stages of craniofacial development. Misexpression in Xenopus of KCNJ2 carrying ATS-associated mutations causes CFAs in the same structures affected in humans, changes the normal pattern of membrane voltage potential regionalization in the developing face and disrupts expression of important craniofacial patterning genes, revealing the endogenous control of craniofacial patterning by bioelectric cell states. By altering cells' resting potentials using other ion translocators, we show that a change in ectodermal voltage, not tied to a specific protein or ion, is sufficient to cause CFAs. By adapting optogenetics for use in non-neural cells in embryos, we show that developmentally patterned K(+) flux is required for correct regionalization of the resting potentials and for establishment of endogenous early gene expression domains in the anterior ectoderm, and that variants in KCNJ2 disrupt this regionalization, leading to the CFAs seen in ATS patients.


Assuntos
Síndrome de Andersen/genética , Anormalidades Craniofaciais/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Animais , Embrião de Mamíferos , Larva , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/anormalidades , Optogenética , RNA Mensageiro/genética , Xenopus laevis
16.
Small ; 12(5): 612-22, 2016 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-26619365

RESUMO

Biochemical gradients are ubiquitous in biology. At the tissue level, they dictate differentiation patterning or cell migration. Recapitulating in vitro the complexity of such concentration profiles with great spatial and dynamic control is crucial in order to understand the underlying mechanisms of biological phenomena. Here, a microfluidic design capable of generating diffusion-driven, simultaneous or sequential, orthogonal linear concentration gradients in a 3D cell-embedded scaffold is described. Formation and stability of the orthogonal gradients are demonstrated by computational and fluorescent dextran-based characterizations. Then, system utility is explored in two biological systems. First, stem cells are subjected to orthogonal gradients of morphogens in order to mimic the localized differentiation of motor neurons in the neural tube. Similarly to in vivo, motor neurons preferentially differentiate in regions of high concentration of retinoic acid and smoothened agonist (acting as sonic hedgehog), in a concentration-dependent fashion. Then, a rotating gradient is applied to HT1080 cancer cells and the change in migration direction is investigated as the cells adapt to a new chemical environment. The response time of ≈4 h is reported. These two examples demonstrate the versatility of this new design that can also prove useful in many applications including tissue engineering and drug screening.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas Analíticas Microfluídicas/métodos , Animais , Diferenciação Celular , Linhagem Celular , Quimiotaxia/efeitos dos fármacos , Cicloexilaminas/farmacologia , Desenho de Equipamento , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Camundongos , Técnicas Analíticas Microfluídicas/instrumentação , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Tubo Neural/citologia , Tubo Neural/embriologia , Tiofenos/farmacologia , Fatores de Tempo , Tretinoína/farmacologia
17.
Lab Chip ; 15(10): 2258-68, 2015 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-25857537

RESUMO

Cardiac tissue engineering aims to recreate functional tissue constructs similar to the structure and function of the native myocardium. To date, in vitro tissue constructs lack the architectural complexity of a vascular network and the precise motor unit control of muscle fibers. Here, we present a method to construct engineered multi-strip cardiac muscle that simulates the bundle-like architecture of the native myocardium. Densely packed primary myocytes and cardiac fibroblasts were co-cultured with optogenetic, non-excitable cells. The resulting 3D syncytium triggered contraction upon localized blue light illumination to selectively activate and pace the multi-strip cardiac muscles, similar to the activity of pacemaker cells. Acting on a single load, we demonstrated graded force production through light-modulated multi-strip recruitment. These results demonstrate an in vitro platform of optogenetic, multi-strip cardiac muscles that can be used in a wide variety of applications, such as drug discovery, tissue engineering, and bio-hybrid robotic systems.


Assuntos
Técnicas de Cultura de Células/métodos , Fibroblastos/metabolismo , Miócitos Cardíacos/citologia , Engenharia Tecidual/métodos , Animais , Fibroblastos/citologia , Células HEK293 , Humanos , Miócitos Cardíacos/metabolismo , Ratos , Ratos Sprague-Dawley
18.
Prog Biophys Mol Biol ; 115(2-3): 279-93, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25175338

RESUMO

The relatively recent development of microfluidic systems with wide-ranging capabilities for generating realistic 2D or 3D systems with single or multiple cell types has given rise to an extensive collection of platform technologies useful in muscle tissue engineering. These new systems are aimed at (i) gaining fundamental understanding of muscle function, (ii) creating functional muscle constructs in vitro, and (iii) utilizing these constructs a variety of applications. Use of microfluidics to control the various stimuli that promote differentiation of multipotent cells into cardiac or skeletal muscle is first discussed. Next, systems that incorporate muscle cells to produce either 2D sheets or 3D tissues of contractile muscle are described with an emphasis on the more recent 3D platforms. These systems are useful for fundamental studies of muscle biology and can also be incorporated into drug screening assays. Applications are discussed for muscle actuators in the context of microrobotics and in miniaturized biological pumps. Finally, an important area of recent study involves coculture with cell types that either activate muscle or facilitate its function. Limitations of current designs and the potential for improving functionality for a wider range of applications is also discussed, with a look toward using current understanding and capabilities to design systems of greater realism, complexity and functionality.


Assuntos
Técnicas Analíticas Microfluídicas/instrumentação , Células Musculares/citologia , Células Musculares/fisiologia , Desenvolvimento Muscular/fisiologia , Engenharia Tecidual/instrumentação , Alicerces Teciduais , Animais , Células Cultivadas , Desenho de Equipamento , Análise de Falha de Equipamento , Humanos , Miniaturização
19.
Laser Phys Lett ; 11(11)2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25620902

RESUMO

A method for selectively inducing apoptosis in tumor nodules is presented, with close-to-cellular level resolution, using 3D-resolved widefield temporal focusing illumination. Treatment times on the order of seconds were achieved using Verteporfin as the photosensitizer, with doses of 30 µg ml-1 and below. Results were achieved on both 2D and 3D cell cultures, demonstrating that treatment was possible through approximately one hundred microns of dense tumor nodules.

20.
Lab Chip ; 13(12): 2252-67, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23649165

RESUMO

Mechanotransduction has been a topic of considerable interest since early studies demonstrated a link between mechanical force and biological response. Until recently, studies of fundamental phenomena were based either on in vivo experiments with limited control or direct access, or on large-scale in vitro studies lacking many of the potentially important physiological factors. With the advent of microfluidics, many of the previous limitations of in vitro testing were eliminated or reduced through greater control or combined functionalities. At the same time, imaging capabilities were tremendously enhanced. In this review, we discuss how microfluidics has transformed the study of mechanotransduction. This is done in the context of the various cell types that exhibit force-induced responses and the new biological insights that have been elucidated. We also discuss new microfluidic studies that could produce even more realistic models of in vivo conditions by combining multiple stimuli or creating a more realistic microenvironment.


Assuntos
Mecanotransdução Celular , Técnicas Analíticas Microfluídicas/métodos , Células Sanguíneas/metabolismo , Células Epiteliais/metabolismo , Humanos , Técnicas Analíticas Microfluídicas/instrumentação , Células Musculares/metabolismo , Células Neoplásicas Circulantes/metabolismo , Resistência ao Cisalhamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...