Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 266
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(23): e2118697119, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35648828

RESUMO

The blood­brain barrier represents a significant challenge for the treatment of high-grade gliomas, and our understanding of drug transport across this critical biointerface remains limited. To advance preclinical therapeutic development for gliomas, there is an urgent need for predictive in vitro models with realistic blood­brain-barrier vasculature. Here, we report a vascularized human glioblastoma multiforme (GBM) model in a microfluidic device that accurately recapitulates brain tumor vasculature with self-assembled endothelial cells, astrocytes, and pericytes to investigate the transport of targeted nanotherapeutics across the blood­brain barrier and into GBM cells. Using modular layer-by-layer assembly, we functionalized the surface of nanoparticles with GBM-targeting motifs to improve trafficking to tumors. We directly compared nanoparticle transport in our in vitro platform with transport across mouse brain capillaries using intravital imaging, validating the ability of the platform to model in vivo blood­brain-barrier transport. We investigated the therapeutic potential of functionalized nanoparticles by encapsulating cisplatin and showed improved efficacy of these GBM-targeted nanoparticles both in vitro and in an in vivo orthotopic xenograft model. Our vascularized GBM model represents a significant biomaterials advance, enabling in-depth investigation of brain tumor vasculature and accelerating the development of targeted nanotherapeutics.


Assuntos
Barreira Hematoencefálica , Neoplasias Encefálicas , Permeabilidade Capilar , Glioblastoma , Nanopartículas , Animais , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Células Endoteliais/metabolismo , Glioblastoma/irrigação sanguínea , Glioblastoma/metabolismo , Humanos , Camundongos , Microfluídica , Nanopartículas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Biol Chem ; 298(5): 101693, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35157851

RESUMO

If a coronary blood vessel is occluded and the neighboring cardiomyocytes deprived of oxygen, subsequent reperfusion of the ischemic tissue can lead to oxidative damage due to excessive generation of reactive oxygen species. Cardiomyocytes and their mitochondria are the main energy producers and consumers of the heart, and their metabolic changes during ischemia seem to be a key driver of reperfusion injury. Here, we hypothesized that tracking changes in cardiomyocyte metabolism, such as oxygen and ATP concentrations, would help in identifying points of metabolic failure during ischemia and reperfusion. To track some of these changes continuously from the onset of ischemia through reperfusion, we developed a system of differential equations representing the chemical reactions involved in the production and consumption of 67 molecular species. This model was validated and used to identify conditions present during periods of critical transition in ischemia and reperfusion that could lead to oxidative damage. These simulations identified a range of oxygen concentrations that lead to reverse mitochondrial electron transport at complex I of the respiratory chain and a spike in mitochondrial membrane potential, which are key suspects in the generation of reactive oxygen species at the onset of reperfusion. Our model predicts that a short initial reperfusion treatment with reduced oxygen content (5% of physiological levels) could reduce the cellular damage from both of these mechanisms. This model should serve as an open-source platform to test ideas for treatment of the ischemia reperfusion process by following the temporal evolution of molecular concentrations in the cardiomyocyte.


Assuntos
Simulação por Computador , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos , Reperfusão/métodos , Humanos , Isquemia/metabolismo , Mitocôndrias Cardíacas/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
3.
FASEB J ; 36(8): e22453, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35838893

RESUMO

Constructing engineered human skeletal muscle tissues that resemble the function and microstructure of human skeletal muscles is key to utilizing them in a variety of applications such as drug development, disease modeling, regenerative medicine, and engineering biological machines. However, current in vitro skeletal muscle tissues are far inferior to native muscles in terms of contractile function and lack essential cues for muscle functions, particularly heterotypic cell-cell interactions between myoblasts, endothelial cells, and fibroblasts. Here, we develop an engineered muscle tissue with a coaxial three-layered tubular structure composed of an inner endothelial cell layer, an endomysium-like layer with fibroblasts in the middle, and an outer skeletal muscle cell layer, similar to the architecture of native skeletal muscles. Engineered skeletal muscle tissues with three spatially organized cell types produced thicker myotubes and lowered Young's modulus through extracellular matrix remodeling, resulting in 43% stronger contractile force. Furthermore, we demonstrated that fibroblasts localized in the endomysium layer induced angiogenic sprouting of endothelial cells into the muscle layer more effectively than fibroblasts homogeneously distributed in the muscle layer. This layered tri-culture system enables a structured spatial configuration of the three main cell types of skeletal muscle and promotes desired paracrine signaling, resulting in improved angiogenesis and increased contractile force. This research offers new insights to efficiently obtain new human skeletal muscle models, transplantable tissues, and actuators for biological machines.


Assuntos
Células Endoteliais , Fibras Musculares Esqueléticas , Fibroblastos , Humanos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Perfusão , Engenharia Tecidual/métodos
4.
Int J Mol Sci ; 24(3)2023 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-36768495

RESUMO

The lack of a conventional lymphatic system that permeates throughout the entire human brain has encouraged the identification and study of alternative clearance routes within the cerebrum. In 2012, the concept of the glymphatic system, a perivascular network that fluidically connects the cerebrospinal fluid to the lymphatic vessels within the meninges via the interstitium, emerged. Although its exact mode of action has not yet been fully characterized, the key underlying processes that govern solute transport and waste clearance have been identified. This review briefly describes the perivascular glial-dependent clearance system and elucidates its fundamental role in neurodegenerative diseases. The current knowledge of the glymphatic system is based almost exclusively on animal-based measurements, but these face certain limitations inherent to in vivo experiments. Recent advances in organ-on-a-chip technology are discussed to demonstrate the technology's ability to provide alternative human-based in vitro research models. Herein, the specific focus is on how current microfluidic-based in vitro models of the neurovascular system and neurodegenerative diseases might be employed to (i) gain a deeper understanding of the role and function of the glymphatic system and (ii) to identify new opportunities for pharmacological intervention.


Assuntos
Sistema Glinfático , Doenças Neurodegenerativas , Animais , Humanos , Sistemas Microfisiológicos , Encéfalo , Sistema Linfático
5.
Adv Funct Mater ; 32(43)2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36569597

RESUMO

Self-organized microvascular networks (MVNs) have become key to the development of many microphysiological models. However, the self-organizing nature of this process combined with variations between types or batches of endothelial cells (ECs) often lead to inconsistency or failure to form functional MVNs. Since interstitial flow (IF) has been reported to play a beneficial role in angiogenesis, vasculogenesis, and 3D capillary morphogenesis, we systematically investigated the role IF plays during neovessel formation in a customized single channel microfluidic chip for which IF has been fully characterized. Compared to static conditions, MVNs formed under IF have higher vessel density and diameters and greater network perfusability. Through a series of inhibitory experiments, we demonstrated that IF treatment improves vasculogenesis by ECs through upregulation of matrix metalloproteinase-2 (MMP-2). We then successfully implemented a novel strategy involving the interplay between IF and MMP-2 inhibitor to regulate morphological parameters of the self-organized MVNs, with vascular permeability and perfusability well maintained. The revealed mechanism and proposed methodology were further validated with a brain MVN model. Our findings and methods have the potential to be widely utilized to boost the development of various organotypic MVNs and could be incorporated into related bioengineering applications where perfusable vasculature is desired.

6.
Nat Mater ; 20(2): 145-155, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33199860

RESUMO

In recent years considerable progress has been made in the development of faithful procedures for the differentiation of human pluripotent stem cells (hPSCs). An important step in this direction has also been the derivation of organoids. This technology generally relies on traditional three-dimensional culture techniques that exploit cell-autonomous self-organization responses of hPSCs with minimal control over the external inputs supplied to the system. The convergence of stem cell biology and bioengineering offers the possibility to provide these stimuli in a controlled fashion, resulting in the development of naturally inspired approaches to overcome major limitations of this nascent technology. Based on the current developments, we emphasize the achievements and ongoing challenges of bringing together hPSC organoid differentiation, bioengineering and ethics. This Review underlines the need for providing engineering solutions to gain control of self-organization and functionality of hPSC-derived organoids. We expect that this knowledge will guide the community to generate higher-grade hPSC-derived organoids for further applications in developmental biology, drug screening, disease modelling and personalized medicine.


Assuntos
Bioengenharia , Organoides/crescimento & desenvolvimento , Células-Tronco Pluripotentes/metabolismo , Humanos , Organoides/citologia , Células-Tronco Pluripotentes/citologia
7.
Annu Rev Biomed Eng ; 23: 359-384, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34255993

RESUMO

The blood-brain barrier (BBB) is one of the most selective endothelial barriers. An understanding of its cellular, morphological, and biological properties in health and disease is necessary to develop therapeutics that can be transported from blood to brain. In vivo models have provided some insight into these features and transport mechanisms adopted at the brain, yet they have failed as a robust platform for the translation of results into clinical outcomes. In this article, we provide a general overview of major BBB features and describe various models that have been designed to replicate this barrier and neurological pathologies linked with the BBB. We propose several key parameters and design characteristics that can be employed to engineer physiologically relevant models of the blood-brain interface and highlight the need for a consensus in the measurement of fundamental properties of this barrier.


Assuntos
Barreira Hematoencefálica , Encéfalo , Transporte Biológico , Biologia , Humanos
8.
Proc Natl Acad Sci U S A ; 116(5): 1543-1548, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30635415

RESUMO

Pumps are critical life-sustaining components for all animals. At the earliest stages of life, the tubular embryonic heart works as a valveless pump capable of generating unidirectional blood flow. Inspired by this elementary pump, we developed an example of a biohybrid valveless pump-bot powered by engineered skeletal muscle. Our pump-bot consists of a soft hydrogel tube connected at both ends to a stiffer polydimethylsiloxane (PDMS) scaffold, creating an impedance mismatch. A contractile muscle ring wraps around the hydrogel tube at an off-center location, squeezing the tube with or without buckling it locally. Cyclic muscle contractions, spontaneous or electrically stimulated, further squeeze the tube, resulting in elastic waves that propagate along the soft tube and get reflected back at the soft/stiff tube boundaries. Asymmetric placement of muscle ring results in a time delay between the wave arrivals, thus establishing a net unidirectional fluid flow irrespective of whether the tube is buckled or not. Flow rates of up to 22.5 µL/min are achieved by the present pump-bot, which are at least three orders of magnitude higher than those from cardiomyocyte-powered valve pumps of similar size. Owning to its simple geometry, robustness, ease of fabrication, and high pumping performance, our pump-bot is particularly well-suited for a wide range of biomedical applications in microfluidics, drug delivery, biomedical devices, cardiovascular pumping system, and more.


Assuntos
Vasos Sanguíneos/fisiologia , Músculo Esquelético/fisiologia , Fluxo Pulsátil/fisiologia , Animais , Linhagem Celular , Camundongos , Contração Muscular/fisiologia , Miócitos Cardíacos/fisiologia , Fluxo Sanguíneo Regional/fisiologia
9.
Angiogenesis ; 24(1): 111-127, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32955682

RESUMO

Angiogenesis plays a key role in the pathology of diseases such as cancer, diabetic retinopathy, and age-related macular degeneration. Understanding the driving forces of endothelial cell migration and organization, as well as the time frame of these processes, can elucidate mechanisms of action of important pathological pathways. Herein, we have developed an organ-specific microfluidic platform recapitulating the in vivo angiogenic microenvironment by co-culturing mouse primary brain endothelial cells with brain pericytes in a three-dimensional (3D) collagen scaffold. As a proof of concept, we show that this model can be used for studying the angiogenic process and further comparing the angiogenic properties between two different common inbred mouse strains, C57BL/6J and 129S1/SvlmJ. We further show that the newly discovered angiogenesis-regulating gene Padi2 promotes angiogenesis through Dll4/Notch1 signaling by an on-chip mechanistic study. Analysis of the interplay between primary endothelial cells and pericytes in a 3D microfluidic environment assists in the elucidation of the angiogenic response.


Assuntos
Engenharia Celular , Microambiente Celular , Células Endoteliais/patologia , Imageamento Tridimensional , Microfluídica , Pericitos/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Separação Celular , Células Cultivadas , Regulação para Baixo , Células Endoteliais/metabolismo , Camundongos Endogâmicos C57BL , Neovascularização Patológica/patologia , Pericitos/metabolismo , Proteína-Arginina Desiminase do Tipo 2/antagonistas & inibidores , Proteína-Arginina Desiminase do Tipo 2/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
10.
Proc Natl Acad Sci U S A ; 115(3): E390-E399, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29295934

RESUMO

Filopodia have a key role in sensing both chemical and mechanical cues in surrounding extracellular matrix (ECM). However, quantitative understanding is still missing in the filopodial mechanosensing of local ECM stiffness, resulting from dynamic interactions between filopodia and the surrounding 3D ECM fibers. Here we present a method for characterizing the stiffness of ECM that is sensed by filopodia based on the theory of elasticity and discrete ECM fiber. We have applied this method to a filopodial mechanosensing model for predicting directed cell migration toward stiffer ECM. This model provides us with a distribution of force and displacement as well as their time rate of changes near the tip of a filopodium when it is bound to the surrounding ECM fibers. Aggregating these effects in each local region of 3D ECM, we express the local ECM stiffness sensed by the cell and explain polarity in the cellular durotaxis mechanism.


Assuntos
Movimento Celular/fisiologia , Simulação por Computador , Matriz Extracelular/fisiologia , Modelos Biológicos , Fenômenos Biomecânicos , Adesão Celular , Citoesqueleto/fisiologia , Elasticidade , Adesões Focais , Pseudópodes
11.
Proc Natl Acad Sci U S A ; 115(16): 4075-4080, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29618614

RESUMO

Animal cells in tissues are supported by biopolymer matrices, which typically exhibit highly nonlinear mechanical properties. While the linear elasticity of the matrix can significantly impact cell mechanics and functionality, it remains largely unknown how cells, in turn, affect the nonlinear mechanics of their surrounding matrix. Here, we show that living contractile cells are able to generate a massive stiffness gradient in three distinct 3D extracellular matrix model systems: collagen, fibrin, and Matrigel. We decipher this remarkable behavior by introducing nonlinear stress inference microscopy (NSIM), a technique to infer stress fields in a 3D matrix from nonlinear microrheology measurements with optical tweezers. Using NSIM and simulations, we reveal large long-ranged cell-generated stresses capable of buckling filaments in the matrix. These stresses give rise to the large spatial extent of the observed cell-induced matrix stiffness gradient, which can provide a mechanism for mechanical communication between cells.


Assuntos
Forma Celular , Proteínas da Matriz Extracelular/química , Matriz Extracelular/ultraestrutura , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Linhagem Celular Tumoral , Colágeno/química , Simulação por Computador , Citocalasina D/farmacologia , Combinação de Medicamentos , Elasticidade , Células Epiteliais/fisiologia , Células Epiteliais/ultraestrutura , Matriz Extracelular/química , Fibrina/química , Humanos , Laminina/química , Modelos Biológicos , Movimento (Física) , Pinças Ópticas , Proteoglicanas/química , Reologia/métodos , Estresse Mecânico
12.
Proc Natl Acad Sci U S A ; 115(27): 7022-7027, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29915060

RESUMO

Systemic inflammation occurring around the course of tumor progression and treatment are often correlated with adverse oncological outcomes. As such, it is suspected that neutrophils, the first line of defense against infection, may play important roles in linking inflammation and metastatic seeding. To decipher the dynamic roles of inflamed neutrophils during hematogenous dissemination, we employ a multiplexed microfluidic model of the human microvasculature enabling physiologically relevant transport of circulating cells combined with real-time, high spatial resolution observation of heterotypic cell-cell interactions. LPS-stimulated neutrophils (PMNs) and tumor cells (TCs) form heterotypic aggregates under flow, and arrest due to both mechanical trapping and neutrophil-endothelial adhesions. Surprisingly, PMNs are not static following aggregation, but exhibit a confined migration pattern near TC-PMN clusters. We discover that PMNs are chemotactically confined by self-secreted IL-8 and tumor-derived CXCL-1, which are immobilized by the endothelial glycocalyx. This results in significant neutrophil sequestration with arrested tumor cells, leading to the spatial localization of neutrophil-derived IL-8, which also contributes to increasing the extravasation potential of adjacent tumor cells through modulation of the endothelial barrier. Strikingly similar migration patterns and extravasation behaviors were also observed in an in vivo zebrafish model upon PMN-tumor cell coinjection into the embryo vasculature. These insights into the temporal dynamics of intravascular tumor-PMN interactions elucidate the mechanisms through which inflamed neutrophils can exert proextravasation effects at the distant metastatic site.


Assuntos
Quimiocina CXCL1/imunologia , Quimiotaxia/imunologia , Interleucina-8/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Neutrófilos/imunologia , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Quimiotaxia/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Técnicas Analíticas Microfluídicas/métodos , Neoplasias/patologia , Neutrófilos/patologia , Peixe-Zebra
13.
Development ; 144(6): 1128-1136, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28174251

RESUMO

Temporal manipulation of the in vitro environment and growth factors can direct differentiation of human pluripotent stem cells into organoids - aggregates with multiple tissue-specific cell types and three-dimensional structure mimicking native organs. A mechanistic understanding of early organoid formation is essential for improving the robustness of these methods, which is necessary prior to use in drug development and regenerative medicine. We investigated intestinal organoid emergence, focusing on measurable parameters of hindgut spheroids, the intermediate step between definitive endoderm and mature organoids. We found that 13% of spheroids were pre-organoids that matured into intestinal organoids. Spheroids varied by several structural parameters: cell number, diameter and morphology. Hypothesizing that diameter and the morphological feature of an inner mass were key parameters for spheroid maturation, we sorted spheroids using an automated micropipette aspiration and release system and monitored the cultures for organoid formation. We discovered that populations of spheroids with a diameter greater than 75 µm and an inner mass are enriched 1.5- and 3.8-fold for pre-organoids, respectively, thus providing rational guidelines towards establishing a robust protocol for high quality intestinal organoids.


Assuntos
Organoides/crescimento & desenvolvimento , Engenharia Tecidual/métodos , Contagem de Células , Tamanho Celular , Células Cultivadas , Sistema Digestório/citologia , Citometria de Fluxo , Humanos , Organoides/citologia , Esferoides Celulares/citologia
14.
Adv Funct Mater ; 30(48)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33692661

RESUMO

Drug discovery and efficacy in cancer treatments are limited by the inability of pre-clinical models to predict successful outcomes in humans. Limitations remain partly due to their lack of a physiologic tumor microenvironment (TME), which plays a considerable role in drug delivery and tumor response to therapy. Chemotherapeutics and immunotherapies rely on transport through the vasculature, via the smallest capillaries and stroma to the tumor, where passive and active transport processes are at play. Here, a 3D vascularized tumor on-chip is used to examine drug delivery in a relevant TME within a large bed of perfusable vasculature. This system demonstrates highly localized pathophysiological effects of two tumor spheroids (Skov3 and A549) which cause significant changes in vessel density and barrier function. Paclitaxel (Taxol) uptake is examined through diffusivity measurements, functional efflux assays and accumulation of the fluorescent-conjugated drug within the TME. Due to vascular and stromal contributions, differences in the response of vascularized tumors to Taxol (shrinkage and CD44 expression) are apparent compared with simpler models. This model specifically allows for examination of spatially resolved tumor-associated endothelial dysfunction, likely improving the representation of in vivo drug distribution, and has potential for development into a more predictable model of drug delivery.

15.
PLoS Comput Biol ; 15(4): e1006684, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30958816

RESUMO

The mechanical properties of the extracellular matrix (ECM)-a complex, 3D, fibrillar scaffold of cells in physiological environments-modulate cell behavior and can drive tissue morphogenesis, regeneration, and disease progression. For simplicity, it is often convenient to assume these properties to be time-invariant. In living systems, however, cells dynamically remodel the ECM and create time-dependent local microenvironments. Here, we show how cell-generated contractile forces produce substantial irreversible changes to the density and architecture of physiologically relevant ECMs-collagen I and fibrin-in a matter of minutes. We measure the 3D deformation profiles of the ECM surrounding cancer and endothelial cells during stages when force generation is active or inactive. We further correlate these ECM measurements to both discrete fiber simulations that incorporate fiber crosslink unbinding kinetics and continuum-scale simulations that account for viscoplastic and damage features. Our findings further confirm that plasticity, as a mechanical law to capture remodeling in these networks, is fundamentally tied to material damage via force-driven unbinding of fiber crosslinks. These results characterize in a multiscale manner the dynamic nature of the mechanical environment of physiologically mimicking cell-in-gel systems.


Assuntos
Matriz Extracelular/fisiologia , Pseudópodes/fisiologia , Fenômenos Biomecânicos , Biopolímeros/química , Biopolímeros/fisiologia , Linhagem Celular , Microambiente Celular/fisiologia , Biologia Computacional , Simulação por Computador , Matriz Extracelular/química , Matriz Extracelular/ultraestrutura , Células Endoteliais da Veia Umbilical Humana , Humanos , Imageamento Tridimensional , Cinética , Modelos Biológicos , Pseudópodes/química , Pseudópodes/ultraestrutura
16.
PLoS Comput Biol ; 15(5): e1006395, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31048903

RESUMO

The formation of gaps in the endothelium is a crucial process underlying both cancer and immune cell extravasation, contributing to the functioning of the immune system during infection, the unfavorable development of chronic inflammation and tumor metastasis. Here, we present a stochastic-mechanical multiscale model of an endothelial cell monolayer and show that the dynamic nature of the endothelium leads to spontaneous gap formation, even without intervention from the transmigrating cells. These gaps preferentially appear at the vertices between three endothelial cells, as opposed to the border between two cells. We quantify the frequency and lifetime of these gaps, and validate our predictions experimentally. Interestingly, we find experimentally that cancer cells also preferentially extravasate at vertices, even when they first arrest on borders. This suggests that extravasating cells, rather than initially signaling to the endothelium, might exploit the autonomously forming gaps in the endothelium to initiate transmigration.


Assuntos
Células Endoteliais/metabolismo , Endotélio Vascular/patologia , Junções Comunicantes/patologia , Fenômenos Biomecânicos , Adesão Celular , Movimento Celular/fisiologia , Células Endoteliais/fisiologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Neoplasias/patologia , Migração Transendotelial e Transepitelial
17.
Adv Exp Med Biol ; 1224: 87-115, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32036607

RESUMO

Monocytes (Mos) are immune cells that critically regulate cancer, enabling tumor growth and modulating metastasis. Mos can give rise to tumor-associated macrophages (TAMs) and Mo-derived dendritic cells (moDCs), all of which shape the tumor microenvironment (TME). Thus, understanding their roles in the TME is key for improved immunotherapy. Concurrently, various biological and mechanical factors including changes in local cytokines, extracellular matrix production, and metabolic changes in the TME affect the roles of monocytic cells. As such, relevant TME models are critical to achieve meaningful insight on the precise functions, mechanisms, and effects of monocytic cells. Notably, murine models have yielded significant insight into human Mo biology. However, many of these results have yet to be confirmed in humans, reinforcing the need for improved in vitro human TME models for the development of cancer interventions. Thus, this chapter (1) summarizes current insight on the tumor biology of Mos, TAMs, and moDCs, (2) highlights key therapeutic applications relevant to these cells, and (3) discusses various TME models to study their TME-related activity. We conclude with a perspective on the future research trajectory of this topic.


Assuntos
Monócitos/patologia , Neoplasias/patologia , Microambiente Tumoral , Animais , Humanos , Imunoterapia , Macrófagos/patologia , Neoplasias/imunologia , Neoplasias/terapia , Microambiente Tumoral/imunologia
18.
Small ; 15(46): e1902393, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31497931

RESUMO

In vitro prediction of physiologically relevant transport of therapeutic molecules across the microcirculation represents an intriguing opportunity to predict efficacy in human populations. On-chip microvascular networks (MVNs) show physiologically relevant values of molecular permeability, yet like most systems, they lack an important contribution to transport: the ever-present fluid convection through the endothelium. Quantification of transport through the MVNs by current methods also requires confocal imaging and advanced analytical techniques, which can be a bottleneck in industry and academic laboratories. Here, it is shown that by recapitulating physiological transmural flow across the MVNs, the concentration of small and large molecule therapeutics can be directly sampled in the interstitial fluid and analyzed using standard analytical techniques. The magnitudes of transport measured in MVNs reveal trends with molecular size and type (protein versus nonprotein) that are expected in vivo, supporting the use of the MVNs platform as an in vitro tool to predict distribution of therapeutics in vivo.


Assuntos
Líquido Extracelular/fisiologia , Microvasos/fisiologia , Fluxo Sanguíneo Regional/fisiologia , Proteínas Sanguíneas/metabolismo , Fluoresceína-5-Isotiocianato/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Dispositivos Lab-On-A-Chip , Perfusão , Permeabilidade , Pressão , Transporte Proteico
19.
Mol Pharm ; 16(6): 2838-2844, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31013423

RESUMO

An optimal response to immune checkpoint blockade requires the presence of effector cells in the tumor microenvironment. We designed a PD-L1-targeted delivery strategy for chemokines, key molecules that drive leukocyte trafficking, to the tumor microenvironment, as a means of attracting the relevant leukocyte populations. This strategy combines a PD-L1-blocking single-domain antibody fragment (nanobody or VHH), a charge-engineered chemokine CCL21, and its subsequent characterization in a microfluidic device that resembles the tumor microenvironment. We show that the PD-L1-blocking VHH is a reliable fusion partner for the preparation of a functional chemokine fusion. Engineering the surface charge of CCL21 reduced its nonspecific binding to glycosaminoglycans, a property of chemokines that complicates their targeted delivery. Using a microfluidic assay, we show that it is possible to deliver a chemokine-VHH fusion to a PD-L1-positive environment and recruit effector cells.


Assuntos
Quimiocinas/metabolismo , Microambiente Tumoral/fisiologia , Animais , Anticorpos Monoclonais/metabolismo , Quimiocina CCL21/metabolismo , Matriz Extracelular/metabolismo , Humanos , Dispositivos Lab-On-A-Chip
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA