Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.833
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(18): 4597-4611, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34478657

RESUMEN

We explore the utility of bioengineered human tissues-individually or connected into physiological units-for biological research. While much smaller and simpler than their native counterparts, these tissues are complex enough to approximate distinct tissue phenotypes: molecular, structural, and functional. Unlike organoids, which form spontaneously and recapitulate development, "organs-on-a-chip" are engineered to display some specific functions of whole organs. Looking back, we discuss the key developments of this emerging technology. Thinking forward, we focus on the challenges faced to fully establish, validate, and utilize the fidelity of these models for biological research.


Asunto(s)
Dispositivos Laboratorio en un Chip , Modelos Biológicos , Investigación , Animales , Ingeniería Celular , Microambiente Celular , Humanos , Ingeniería de Tejidos
2.
Annu Rev Biochem ; 86: 333-356, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28654324

RESUMEN

Many biochemical systems are spatially heterogeneous and exhibit nonlinear behaviors, such as state switching in response to small changes in the local concentration of diffusible molecules. Systems as varied as blood clotting, intracellular calcium signaling, and tissue inflammation are all heavily influenced by the balance of rates of reaction and mass transport phenomena including flow and diffusion. Transport of signaling molecules is also affected by geometry and chemoselective confinement via matrix binding. In this review, we use a phenomenon referred to as patchy switching to illustrate the interplay of nonlinearities, transport phenomena, and spatial effects. Patchy switching describes a change in the state of a network when the local concentration of a diffusible molecule surpasses a critical threshold. Using patchy switching as an example, we describe conceptual tools from nonlinear dynamics and chemical engineering that make testable predictions and provide a unifying description of the myriad possible experimental observations. We describe experimental microfluidic and biochemical tools emerging to test conceptual predictions by controlling transport phenomena and spatial distribution of diffusible signals, and we highlight the unmet need for in vivo tools.


Asunto(s)
Adenocarcinoma/metabolismo , Redes Reguladoras de Genes , Neoplasias Pulmonares/metabolismo , Redes y Vías Metabólicas/genética , Esclerosis Múltiple/metabolismo , Dinámicas no Lineales , Osteoporosis/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Transporte Biológico , Difusión , Humanos , Dispositivos Laboratorio en un Chip , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Microfluídica/instrumentación , Microfluídica/métodos , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Osteoporosis/genética , Osteoporosis/patología , Transducción de Señal
3.
Nat Rev Genet ; 23(8): 467-491, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35338360

RESUMEN

The failure of animal models to predict therapeutic responses in humans is a major problem that also brings into question their use for basic research. Organ-on-a-chip (organ chip) microfluidic devices lined with living cells cultured under fluid flow can recapitulate organ-level physiology and pathophysiology with high fidelity. Here, I review how single and multiple human organ chip systems have been used to model complex diseases and rare genetic disorders, to study host-microbiome interactions, to recapitulate whole-body inter-organ physiology and to reproduce human clinical responses to drugs, radiation, toxins and infectious pathogens. I also address the challenges that must be overcome for organ chips to be accepted by the pharmaceutical industry and regulatory agencies, as well as discuss recent advances in the field. It is evident that the use of human organ chips instead of animal models for drug development and as living avatars for personalized medicine is ever closer to realization.


Asunto(s)
Dispositivos Laboratorio en un Chip , Medicina de Precisión , Animales , Desarrollo de Medicamentos , Humanos , Enfermedades Raras
4.
Nature ; 605(7910): 464-469, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35585345

RESUMEN

Chain reactions, characterized by initiation, propagation and termination, are stochastic at microscopic scales and underlie vital chemical (for example, combustion engines), nuclear and biotechnological (for example, polymerase chain reaction) applications1-5. At macroscopic scales, chain reactions are deterministic and limited to applications for entertainment and art such as falling dominoes and Rube Goldberg machines. On the other hand, the microfluidic lab-on-a-chip (also called a micro-total analysis system)6,7 was visualized as an integrated chip, akin to microelectronic integrated circuits, yet in practice remains dependent on cumbersome peripherals, connections and a computer for automation8-11. Capillary microfluidics integrate energy supply and flow control onto a single chip by using capillary phenomena, but programmability remains rudimentary with at most a handful (eight) operations possible12-19. Here we introduce the microfluidic chain reaction (MCR) as the conditional, structurally programmed propagation of capillary flow events. Monolithic chips integrating a MCR are three-dimensionally printed, and powered by the free energy of a paper pump, autonomously execute liquid handling algorithms step-by-step. With MCR, we automated (1) the sequential release of 300 aliquots across chained, interconnected chips, (2) a protocol for severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) antibodies detection in saliva and (3) a thrombin generation assay by continuous subsampling and analysis of coagulation-activated plasma with parallel operations including timers, iterative cycles of synchronous flow and stop-flow operations. MCRs are untethered from and unencumbered by peripherals, encode programs structurally in situ and can form a frugal, versatile, bona fide lab-on-a-chip with wide-ranging applications in liquid handling and point-of-care diagnostics.


Asunto(s)
COVID-19 , Técnicas Analíticas Microfluídicas , Humanos , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/métodos , Microfluídica/métodos , Reacción en Cadena de la Polimerasa , SARS-CoV-2/genética
5.
PLoS Biol ; 22(3): e3002503, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38478490

RESUMEN

Cell culture devices, such as microwells and microfluidic chips, are designed to increase the complexity of cell-based models while retaining control over culture conditions and have become indispensable platforms for biological systems modelling. From microtopography, microwells, plating devices, and microfluidic systems to larger constructs such as live imaging chamber slides, a wide variety of culture devices with different geometries have become indispensable in biology laboratories. However, while their application in biological projects is increasing exponentially, due to a combination of the techniques, equipment and tools required for their manufacture, and the expertise necessary, biological and biomedical labs tend more often to rely on already made devices. Indeed, commercially developed devices are available for a variety of applications but are often costly and, importantly, lack the potential for customisation by each individual lab. The last point is quite crucial, as often experiments in wet labs are adapted to whichever design is already available rather than designing and fabricating custom systems that perfectly fit the biological question. This combination of factors still restricts widespread application of microfabricated custom devices in most biological wet labs. Capitalising on recent advances in bioengineering and microfabrication aimed at solving these issues, and taking advantage of low-cost, high-resolution desktop resin 3D printers combined with PDMS soft lithography, we have developed an optimised a low-cost and highly reproducible microfabrication pipeline. This is thought specifically for biomedical and biological wet labs with not prior experience in the field, which will enable them to generate a wide variety of customisable devices for cell culture and tissue engineering in an easy, fast reproducible way for a fraction of the cost of conventional microfabrication or commercial alternatives. This protocol is designed specifically to be a resource for biological labs with limited expertise in those techniques and enables the manufacture of complex devices across the µm to cm scale. We provide a ready-to-go pipeline for the efficient treatment of resin-based 3D-printed constructs for PDMS curing, using a combination of polymerisation steps, washes, and surface treatments. Together with the extensive characterisation of the fabrication pipeline, we show the utilisation of this system to a variety of applications and use cases relevant to biological experiments, ranging from micro topographies for cell alignments to complex multipart hydrogel culturing systems. This methodology can be easily adopted by any wet lab, irrespective of prior expertise or resource availability and will enable the wide adoption of tailored microfabricated devices across many fields of biology.


Asunto(s)
Técnicas de Cultivo de Célula , Microtecnología , Microfluídica/métodos , Impresión Tridimensional , Dispositivos Laboratorio en un Chip
6.
Proc Natl Acad Sci U S A ; 121(11): e2316500121, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38442157

RESUMEN

Evaluating the ability of cytotoxic T lymphocytes (CTLs) to eliminate tumor cells is crucial, for instance, to predict the efficiency of cell therapy in personalized medicine. However, the destruction of a tumor by CTLs involves CTL migration in the extra-tumoral environment, accumulation on the tumor, antigen recognition, and cooperation in killing the cancer cells. Therefore, identifying the limiting steps in this complex process requires spatio-temporal measurements of different cellular events over long periods. Here, we use a cancer-on-a-chip platform to evaluate the impact of adenomatous polyposis coli (APC) mutation on CTL migration and cytotoxicity against 3D tumor spheroids. The APC mutated CTLs are found to have a reduced ability to destroy tumor spheroids compared with control cells, even though APC mutants migrate in the extra-tumoral space and accumulate on the spheroids as efficiently as control cells. Once in contact with the tumor however, mutated CTLs display reduced engagement with the cancer cells, as measured by a metric that distinguishes different modes of CTL migration. Realigning the CTL trajectories around localized killing cascades reveals that all CTLs transition to high engagement in the 2 h preceding the cascades, which confirms that the low engagement is the cause of reduced cytotoxicity. Beyond the study of APC mutations, this platform offers a robust way to compare cytotoxic cell efficiency of even closely related cell types, by relying on a multiscale cytometry approach to disentangle complex interactions and to identify the steps that limit the tumor destruction.


Asunto(s)
Poliposis Adenomatosa del Colon , Neoplasias , Humanos , Neoplasias/genética , Linfocitos T Citotóxicos , Mutación , Dispositivos Laboratorio en un Chip
7.
Proc Natl Acad Sci U S A ; 121(28): e2403581121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38968108

RESUMEN

Adverse cardiac outcomes in COVID-19 patients, particularly those with preexisting cardiac disease, motivate the development of human cell-based organ-on-a-chip models to recapitulate cardiac injury and dysfunction and for screening of cardioprotective therapeutics. Here, we developed a heart-on-a-chip model to study the pathogenesis of SARS-CoV-2 in healthy myocardium established from human induced pluripotent stem cell (iPSC)-derived cardiomyocytes and a cardiac dysfunction model, mimicking aspects of preexisting hypertensive disease induced by angiotensin II (Ang II). We recapitulated cytopathic features of SARS-CoV-2-induced cardiac damage, including progressively impaired contractile function and calcium handling, apoptosis, and sarcomere disarray. SARS-CoV-2 presence in Ang II-treated hearts-on-a-chip decreased contractile force with earlier onset of contractile dysfunction and profoundly enhanced inflammatory cytokines compared to SARS-CoV-2 alone. Toward the development of potential therapeutics, we evaluated the cardioprotective effects of extracellular vesicles (EVs) from human iPSC which alleviated the impairment of contractile force, decreased apoptosis, reduced the disruption of sarcomeric proteins, and enhanced beta-oxidation gene expression. Viral load was not affected by either Ang II or EV treatment. We identified MicroRNAs miR-20a-5p and miR-19a-3p as potential mediators of cardioprotective effects of these EVs.


Asunto(s)
Angiotensina II , COVID-19 , Vesículas Extracelulares , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , SARS-CoV-2 , Humanos , Angiotensina II/farmacología , COVID-19/virología , COVID-19/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/virología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Vesículas Extracelulares/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Apoptosis/efectos de los fármacos , Dispositivos Laboratorio en un Chip , MicroARNs/metabolismo , MicroARNs/genética , Citocinas/metabolismo
8.
Proc Natl Acad Sci U S A ; 120(32): e2115616120, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37494421

RESUMEN

Transfusion of red blood cells (RBCs) is one of the most valuable and widespread treatments in modern medicine. Lifesaving RBC transfusions are facilitated by the cold storage of RBC units in blood banks worldwide. Currently, RBC storage and subsequent transfusion practices are performed using simplistic workflows. More specifically, most blood banks follow the "first-in-first-out" principle to avoid wastage, whereas most healthcare providers prefer the "last-in-first-out" approach simply favoring chronologically younger RBCs. Neither approach addresses recent advances through -omics showing that stored RBC quality is highly variable depending on donor-, time-, and processing-specific factors. Thus, it is time to rethink our workflows in transfusion medicine taking advantage of novel technologies to perform RBC quality assessment. We imagine a future where lab-on-a-chip technologies utilize novel predictive markers of RBC quality identified by -omics and machine learning to usher in a new era of safer and precise transfusion medicine.


Asunto(s)
Conservación de la Sangre , Procedimientos Analíticos en Microchip , Transfusión Sanguínea/instrumentación , Transfusión Sanguínea/métodos , Humanos , Conservación de la Sangre/métodos , Dispositivos Laboratorio en un Chip , Eritrocitos , Aprendizaje Automático
9.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35156682

RESUMEN

The proper development and patterning of organs rely on concerted signaling events emanating from intracellular and extracellular molecular and biophysical cues. The ability to model and understand how these microenvironmental factors contribute to cell fate decisions and physiological processes is crucial for uncovering the biology and mechanisms of life. Recent advances in microfluidic systems have provided novel tools and strategies for studying aspects of human tissue and organ development in ways that have previously been challenging to explore ex vivo. Here, we discuss how microfluidic systems and organs-on-chips provide new ways to understand how extracellular signals affect cell differentiation, how cells interact with each other, and how different tissues and organs are formed for specialized functions. We also highlight key advancements in the field that are contributing to a broad understanding of human embryogenesis, organogenesis and physiology. We conclude by summarizing the key advantages of using dynamic microfluidic or microphysiological platforms to study intricate developmental processes that cannot be accurately modeled by using traditional tissue culture vessels. We also suggest some exciting prospects and potential future applications of these emerging technologies.


Asunto(s)
Microfluídica/métodos , Modelos Biológicos , Corazón/crecimiento & desarrollo , Corazón/fisiología , Humanos , Dispositivos Laboratorio en un Chip , Microfluídica/instrumentación , Poliésteres/química , Impresión Tridimensional , Ingeniería de Tejidos
10.
Annu Rev Biomed Eng ; 26(1): 119-139, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38316063

RESUMEN

Recent advances in single-cell and multicellular microfluidics technology have provided powerful tools for studying cancer biology and immunology. The ability to create controlled microenvironments, perform high-throughput screenings, and monitor cellular interactions at the single-cell level has significantly advanced our understanding of tumor biology and immune responses. We discuss cutting-edge multicellular and single-cell microfluidic technologies and methodologies utilized to investigate cancer-immune cell interactions and assess the effectiveness of immunotherapies. We explore the advantages and limitations of the wide range of 3D spheroid and single-cell microfluidic models recently developed, highlighting the various approaches in device generation and applications in immunotherapy screening for potential opportunities for point-of-care approaches.


Asunto(s)
Microfluídica , Neoplasias , Sistemas de Atención de Punto , Análisis de la Célula Individual , Humanos , Análisis de la Célula Individual/métodos , Microfluídica/métodos , Microambiente Tumoral , Inmunoterapia/métodos , Esferoides Celulares , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Comunicación Celular , Animales , Dispositivos Laboratorio en un Chip
11.
Annu Rev Biomed Eng ; 26(1): 383-414, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38424088

RESUMEN

Kidney disease is a global health crisis affecting more than 850 million people worldwide. In the United States, annual Medicare expenditures for kidney disease and organ failure exceed $81 billion. Efforts to develop targeted therapeutics are limited by a poor understanding of the molecular mechanisms underlying human kidney disease onset and progression. Additionally, 90% of drug candidates fail in human clinical trials, often due to toxicity and efficacy not accurately predicted in animal models. The advent of ex vivo kidney models, such as those engineered from induced pluripotent stem (iPS) cells and organ-on-a-chip (organ-chip) systems, has garnered considerable interest owing to their ability to more accurately model tissue development and patient-specific responses and drug toxicity. This review describes recent advances in developing kidney organoids and organ-chips by harnessing iPS cell biology to model human-specific kidney functions and disease states. We also discuss challenges that must be overcome to realize the potential of organoids and organ-chips as dynamic and functional conduits of the human kidney. Achieving these technological advances could revolutionize personalized medicine applications and therapeutic discovery for kidney disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Renales , Riñón , Dispositivos Laboratorio en un Chip , Organoides , Ingeniería de Tejidos , Humanos , Animales , Células Madre Pluripotentes Inducidas/citología , Ingeniería de Tejidos/métodos , Modelos Biológicos , Medicina de Precisión/métodos
12.
Circ Res ; 132(4): 483-497, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36795846

RESUMEN

Heart disease is a significant burden on global health care systems and is a leading cause of death each year. To improve our understanding of heart disease, high quality disease models are needed. These will facilitate the discovery and development of new treatments for heart disease. Traditionally, researchers have relied on 2D monolayer systems or animal models of heart disease to elucidate pathophysiology and drug responses. Heart-on-a-chip (HOC) technology is an emerging field where cardiomyocytes among other cell types in the heart can be used to generate functional, beating cardiac microtissues that recapitulate many features of the human heart. HOC models are showing great promise as disease modeling platforms and are poised to serve as important tools in the drug development pipeline. By leveraging advances in human pluripotent stem cell-derived cardiomyocyte biology and microfabrication technology, diseased HOCs are highly tuneable and can be generated via different approaches such as: using cells with defined genetic backgrounds (patient-derived cells), adding small molecules, modifying the cells' environment, altering cell ratio/composition of microtissues, among others. HOCs have been used to faithfully model aspects of arrhythmia, fibrosis, infection, cardiomyopathies, and ischemia, to name a few. In this review, we highlight recent advances in disease modeling using HOC systems, describing instances where these models outperformed other models in terms of reproducing disease phenotypes and/or led to drug development.


Asunto(s)
Cardiomiopatías , Cardiopatías , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Humanos , Cardiopatías/terapia , Cardiopatías/metabolismo , Miocitos Cardíacos/metabolismo , Cardiomiopatías/metabolismo , Células Madre Pluripotentes/metabolismo , Dispositivos Laboratorio en un Chip
13.
Cell Mol Life Sci ; 81(1): 197, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38664263

RESUMEN

Congenital heart defects are associated with significant health challenges, demanding a deep understanding of the underlying biological mechanisms and, thus, better devices or platforms that can recapitulate human cardiac development. The discovery of human pluripotent stem cells has substantially reduced the dependence on animal models. Recent advances in stem cell biology, genetic editing, omics, microfluidics, and sensor technologies have further enabled remarkable progress in the development of in vitro platforms with increased fidelity and efficiency. In this review, we provide an overview of advancements in in vitro cardiac development platforms, with a particular focus on technological innovation. We categorize these platforms into four areas: two-dimensional solid substrate cultures, engineered substrate architectures that enhance cellular functions, cardiac organoids, and embryos/explants-on-chip models. We conclude by addressing current limitations and presenting future perspectives.


Asunto(s)
Evaluación Preclínica de Medicamentos , Corazón , Ingeniería de Tejidos , Humanos , Animales , Evaluación Preclínica de Medicamentos/métodos , Ingeniería de Tejidos/métodos , Organoides/metabolismo , Organoides/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Cardiopatías Congénitas/genética , Dispositivos Laboratorio en un Chip
14.
Proc Natl Acad Sci U S A ; 119(13): e2115276119, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35312360

RESUMEN

SignificanceThe treatment of hypoxemia that is refractory to the current standard of care is time-sensitive and requires skilled caregivers and use of specialized equipment (e.g., extracorporeal membrane oxygenation). Most patients experiencing refractory hypoxemia will suffer organ dysfunction, and death is common in this cohort. Here, we describe a new strategy to stabilize and support patients using a microfluidic device that administers oxygen gas directly to the bloodstream in real time and on demand using a process that we call sequential shear-induced bubble breakup. If successful, the described technology may help to avoid or decrease the incidence of ventilator-related lung injury from refractory hypoxemia.


Asunto(s)
Oxigenación por Membrana Extracorpórea , Lesión Pulmonar , Oxigenación por Membrana Extracorpórea/efectos adversos , Humanos , Hipoxia , Dispositivos Laboratorio en un Chip , Oxígeno , Ventiladores Mecánicos/efectos adversos
15.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35145031

RESUMEN

Bacteria are efficient colonizers of a wide range of secluded microhabitats, such as soil pores, skin follicles, or intestinal crypts. How the structural diversity of these habitats modulates microbial self-organization remains poorly understood, in part because of the difficulty to precisely manipulate the physical structure of microbial environments. Using a microfluidic device to grow bacteria in crypt-like incubation chambers of systematically varied lengths, we show that small variations in the physical structure of the microhabitat can drastically alter bacterial colonization success and resistance against invaders. Small crypts are uncolonizable; intermediately sized crypts can stably support dilute populations, while beyond a second critical length scale, populations phase separate into a dilute region and a jammed region. The jammed state is characterized by extreme colonization resistance, even if the resident strain is suppressed by an antibiotic. Combined with a flexible biophysical model, we demonstrate that colonization resistance and associated priority effects can be explained by a crowding-induced phase transition, which results from a competition between proliferation and density-dependent cell leakage. The emerging sensitivity to scale underscores the need to control for scale in microbial ecology experiments. Systematic flow-adjustable length-scale variations may serve as a promising strategy to elucidate further scale-sensitive tipping points and to rationally modulate the stability and resilience of microbial colonizers.


Asunto(s)
Acetobacter/fisiología , Dispositivos Laboratorio en un Chip , Acetobacter/efectos de los fármacos , Antibacterianos/farmacología , Técnicas Bacteriológicas , Farmacorresistencia Bacteriana , Tetraciclina/farmacología
16.
Proc Natl Acad Sci U S A ; 119(5)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35074874

RESUMEN

For nearly 50 years, the vision of using single molecules in circuits has been seen as providing the ultimate miniaturization of electronic chips. An advanced example of such a molecular electronics chip is presented here, with the important distinction that the molecular circuit elements play the role of general-purpose single-molecule sensors. The device consists of a semiconductor chip with a scalable array architecture. Each array element contains a synthetic molecular wire assembled to span nanoelectrodes in a current monitoring circuit. A central conjugation site is used to attach a single probe molecule that defines the target of the sensor. The chip digitizes the resulting picoamp-scale current-versus-time readout from each sensor element of the array at a rate of 1,000 frames per second. This provides detailed electrical signatures of the single-molecule interactions between the probe and targets present in a solution-phase test sample. This platform is used to measure the interaction kinetics of single molecules, without the use of labels, in a massively parallel fashion. To demonstrate broad applicability, examples are shown for probe molecule binding, including DNA oligos, aptamers, antibodies, and antigens, and the activity of enzymes relevant to diagnostics and sequencing, including a CRISPR/Cas enzyme binding a target DNA, and a DNA polymerase enzyme incorporating nucleotides as it copies a DNA template. All of these applications are accomplished with high sensitivity and resolution, on a manufacturable, scalable, all-electronic semiconductor chip device, thereby bringing the power of modern chips to these diverse areas of biosensing.


Asunto(s)
Técnicas Biosensibles/instrumentación , Electrónica/instrumentación , Pruebas de Enzimas/instrumentación , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , ADN , Diseño de Equipo/instrumentación , Cinética , Dispositivos Laboratorio en un Chip , Miniaturización/instrumentación , Nanotecnología/instrumentación , Semiconductores
17.
Nano Lett ; 24(26): 8151-8161, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38912914

RESUMEN

The size of liposomal drugs has been demonstrated to strongly correlate with their pharmacokinetics and pharmacodynamics. While the microfluidic method successfully achieves the production of liposomes with well-controlled sizes across various buffer/lipid flow rate ratio (FRR) settings, any adjustments to the FRR inevitably influence the concentration, encapsulation efficiency (EE), and stability of liposomal drugs. Here we describe a controllable cavitation-on-a-chip (CCC) strategy that facilitates the precise regulation of liposomal drug size at any desired FRR. The CCC-enabled size-specific liposomes exhibited striking differences in uptake and biodistribution behaviors, thereby demonstrating distinct antitumor efficacy in both tumor-bearing animal and melanoma patient-derived organoid (PDO) models. Intriguingly, as the liposome size decreased to approximately 80 nm, the preferential accumulation of liposomal drugs in the liver transitioned to a predominant enrichment in the kidneys. These findings underscore the considerable potential of our CCC approach in influencing the pharmacokinetics and pharmacodynamics of liposomal nanomedicines.


Asunto(s)
Dispositivos Laboratorio en un Chip , Liposomas , Liposomas/química , Animales , Humanos , Ratones , Distribución Tisular , Tamaño de la Partícula , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Melanoma/tratamiento farmacológico , Melanoma/patología
18.
Kidney Int ; 105(4): 702-708, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38296026

RESUMEN

The use of animal models in therapeutic development has long been the standard practice. However, ethical concerns and the inherent species differences have prompted a reevaluation of the experimental approach in human disease studies. The urgent need for alternative model systems that better mimic human pathophysiology has led to the emergence of organoids, innovative in vitro models, to simulate human organs in vitro. These organoids have gained widespread acceptance in disease models and drug development research. In this mini review, we explore the recent strides made in kidney organoid differentiation and highlight the synergistic potential of incorporating organ-on-chip systems. The emergent use of microfluidic devices reveals the importance of fluid flow in the maturation of kidney organoids and helps decipher pathomechanisms in kidney diseases. Recent research has uncovered their potential applications across a wide spectrum of kidney research areas, including hemodynamic forces at stake in kidney health and disease, immune cell infiltration, or drug delivery and toxicity. This convergence of cutting-edge technologies not only holds promise for expediting therapeutic development but also reflects an acknowledgment of the need to embrace innovative and more human-centric research models.


Asunto(s)
Riñón , Organoides , Animales , Diferenciación Celular , Sistemas de Liberación de Medicamentos , Dispositivos Laboratorio en un Chip
19.
Angiogenesis ; 27(2): 147-172, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38409567

RESUMEN

Vascularized organoid-on-a-chip (VOoC) models achieve substance exchange in deep layers of organoids and provide a more physiologically relevant system in vitro. Common designs for VOoC primarily involve two categories: self-assembly of endothelial cells (ECs) to form microvessels and pre-patterned vessel lumens, both of which include the hydrogel region for EC growth and allow for controlled fluid perfusion on the chip. Characterizing the vasculature of VOoC often relies on high-resolution microscopic imaging. However, the high scattering of turbid tissues can limit optical imaging depth. To overcome this limitation, tissue optical clearing (TOC) techniques have emerged, allowing for 3D visualization of VOoC in conjunction with optical imaging techniques. The acquisition of large-scale imaging data, coupled with high-resolution imaging in whole-mount preparations, necessitates the development of highly efficient analysis methods. In this review, we provide an overview of the chip designs and culturing strategies employed for VOoC, as well as the applicable optical imaging and TOC methods. Furthermore, we summarize the vascular analysis techniques employed in VOoC, including deep learning. Finally, we discuss the existing challenges in VOoC and vascular analysis methods and provide an outlook for future development.


Asunto(s)
Células Endoteliales , Organoides , Hidrogeles , Microvasos , Dispositivos Laboratorio en un Chip
20.
Anal Chem ; 96(28): 11412-11421, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38954777

RESUMEN

Efficient cell manipulation is essential for numerous applications in bioanalysis and medical diagnosis. However, the lack of stability and strength in the secondary flow, coupled with the narrow range of practical throughput, severely restricts the diverse applications. Herein, we present an innovative inertial microfluidic device that employs a spiral channel for high-throughput cell manipulation. Our investigation demonstrates that the regulation of Dean-like secondary flow in the microchannel can be achieved through geometric confinement. Introducing ordered microstructures into the ultralong spiral channel (>90 cm) stabilizes and accelerates the secondary flow among different loops. Consequently, effective manipulation of blood cells within a wide cell throughput range (1.73 × 108 to 1.16 × 109 cells/min) and cancer cells across a broad throughput range (0.5 × 106 to 5 × 107 cells/min) can be achieved. In comparison to previously reported technologies, our engineering approach of stabilizing and accelerating secondary flow offers specific performance for cell manipulation under a wide range of high-throughput manner. This engineered spiral channel would be promising in biomedical analysis, especially when cells need to be focused efficiently on large-volume liquid samples.


Asunto(s)
Dispositivos Laboratorio en un Chip , Humanos , Técnicas Analíticas Microfluídicas/instrumentación , Ensayos Analíticos de Alto Rendimiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA