Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Adv Healthc Mater ; : e2303419, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38686434

RESUMO

Microvessels, including arterioles, capillaries, and venules, play an important role in regulating blood flow, enabling nutrient and waste exchange, and facilitating immune surveillance. Due to their important roles in maintaining normal function in human tissues, a substantial effort has been devoted to developing tissue-engineered models to study endothelium-related biology and pathology. Various engineering strategies have been developed to recapitulate the structural, cellular, and molecular hallmarks of native human microvessels in vitro. In this review, recent progress in engineering approaches, key components, and culture platforms for tissue-engineered human microvessel models is summarized. Then, tissue-specific models, and the major applications of tissue-engineered microvessels in development, disease modeling, drug screening and delivery, and vascularization in tissue engineering, are reviewed. Finally, future research directions for the field are discussed.

2.
J Med Chem ; 66(24): 16615-16627, 2023 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-38097510

RESUMO

We report molecular dynamics simulations of rhodamine entry into the central binding cavity of P-gp in the inward open conformation. Rhodamine can enter the inner volume via passive transport across the luminal membrane or lateral diffusion in the lipid bilayer. Entry into the inner volume is determined by the aperture angle at the apex of the protein, with a critical angle of 27° for rhodamine. The central binding cavity has an aqueous phase with a few lipids, which significantly reduces substrate diffusion. Within the central binding cavity, we identified regions with relatively weak binding, suggesting that the combination of reduced mobility and weak substrate binding confines rhodamine to enable the completion of the efflux cycle. Tariquidar, a P-gp inhibitor, aggregates at the lower arms of the P-gp, suggesting that inhibition involves steric hindrance of entry into the inner volume and/or steric hindrance of access of ATP to the nucleotide-binding domains.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Barreira Hematoencefálica , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Barreira Hematoencefálica/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Transporte Biológico , Rodaminas
3.
Fluids Barriers CNS ; 20(1): 80, 2023 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-37924145

RESUMO

Metastatic brain cancer has poor prognosis due to challenges in both detection and treatment. One contributor to poor prognosis is the blood-brain barrier (BBB), which severely limits the transport of therapeutic agents to intracranial tumors. During the development of brain metastases from primary breast cancer, the BBB is modified and is termed the 'blood-tumor barrier' (BTB). A better understanding of the differences between the BBB and BTB across cancer types and stages may assist in identifying new therapeutic targets. Here, we utilize a tissue-engineered microvessel model with induced pluripotent stem cell (iPSC)-derived brain microvascular endothelial-like cells (iBMECs) and surrounded by human breast metastatic cancer spheroids with brain tropism. We directly compare BBB and BTB in vitro microvessels to unravel both physical and chemical interactions occurring during perivascular cancer growth. We determine the dynamics of vascular co-option by cancer cells, modes of vascular degeneration, and quantify the endothelial barrier to antibody transport. Additionally, using bulk RNA sequencing, ELISA of microvessel perfusates, and related functional assays, we probe early brain endothelial changes in the presence of cancer cells. We find that immune cell adhesion and endothelial turnover are elevated within the metastatic BTB, and that macrophages exert a unique influence on BTB identity. Our model provides a novel three-dimensional system to study mechanisms of cancer-vascular-immune interactions and drug delivery occurring within the BTB.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Células-Tronco Pluripotentes Induzidas , Humanos , Feminino , Neoplasias da Mama/patologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Neoplasias Encefálicas/metabolismo , Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo
4.
Front Neurosci ; 17: 1289894, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37937070

RESUMO

The blood-brain barrier (BBB) is located at the interface between the vascular system and the brain parenchyma, and is responsible for communication with systemic circulation and peripheral tissues. During life, the BBB can be subjected to a wide range of perturbations or stresses that may be endogenous or exogenous, pathological or therapeutic, or intended or unintended. The risk factors for many diseases of the brain are multifactorial and involve perturbations that may occur simultaneously (e.g., two-hit model for Alzheimer's disease) and result in different outcomes. Therefore, it is important to understand the influence of individual perturbations on BBB function in isolation. Here we review the effects of eight perturbations: mechanical forces, temperature, electromagnetic radiation, hypoxia, endogenous factors, exogenous factors, chemical factors, and pathogens. While some perturbations may result in acute or chronic BBB disruption, many are also exploited for diagnostic or therapeutic purposes. The resultant outcome on BBB function depends on the dose (or magnitude) and duration of the perturbation. Homeostasis may be restored by self-repair, for example, via processes such as proliferation of affected cells or angiogenesis to create new vasculature. Transient or sustained BBB dysfunction may result in acute or pathological symptoms, for example, microhemorrhages or hypoperfusion. In more extreme cases, perturbations may lead to cytotoxicity and cell death, for example, through exposure to cytotoxic plaques.

5.
J Comput Aided Mol Des ; 37(11): 537-549, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37573260

RESUMO

The treatment of various disorders of the central nervous system (CNS) is often impeded by the limited brain exposure of drugs, which is regulated by the human blood-brain barrier (BBB). The screening of lead compounds for CNS penetration is challenging due to the biochemical complexity of the BBB, while experimental determination of permeability is not feasible for all types of compounds. Here we present a novel method for rapid preclinical screening of libraries of compounds by utilizing advancements in computing hardware, with its foundation in transition-based counting of the flux. This method has been experimentally validated for in vitro permeabilities and provides atomic-level insights into transport mechanisms. Our approach only requires a single high-temperature simulation to rank a compound relative to a library, with a typical simulation time converging within 24 to 72 h. The method offers unbiased thermodynamic and kinetic information to interpret the passive transport of small-molecule drugs across the BBB.


Assuntos
Barreira Hematoencefálica , Humanos , Transporte Biológico/fisiologia , Permeabilidade , Simulação por Computador , Endotélio
8.
NPJ Digit Med ; 5(1): 164, 2022 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-36352062

RESUMO

Physical health status defines an individual's ability to perform normal activities of daily living and is usually assessed in clinical settings by questionnaires and/or by validated tests, e.g. timed walk tests. These measurements have relatively low information content and are usually limited in frequency. Wearable sensors, such as activity monitors, enable remote measurement of parameters associated with physical activity but have not been widely explored beyond measurement of daily step count. Here we report on results from a cohort of 22 individuals with Pulmonary Arterial Hypertension (PAH) who were provided with a Fitbit activity monitor (Fitbit Charge HR®) between two clinic visits (18.4 ± 12.2 weeks). At each clinical visit, a maximum of 26 measurements were recorded (19 categorical and 7 continuous). From analysis of the minute-to-minute step rate and heart rate we derive several metrics associated with physical activity and cardiovascular function. These metrics are used to identify subgroups within the cohort and to compare to clinical parameters. Several Fitbit metrics are strongly correlated to continuous clinical parameters. Using a thresholding approach, we show that many Fitbit metrics result in statistically significant differences in clinical parameters between subgroups, including those associated with physical status, cardiovascular function, pulmonary function, as well as biomarkers from blood tests. These results highlight the fact that daily step count is only one of many metrics that can be derived from activity monitors.

9.
Fluids Barriers CNS ; 19(1): 87, 2022 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-36333694

RESUMO

The blood-brain barrier (BBB) plays a pivotal role in brain health and disease. In the BBB, brain microvascular endothelial cells (BMECs) are connected by tight junctions which regulate paracellular transport, and express specialized transporter systems which regulate transcellular transport. However, existing in vitro models of the BBB display variable accuracy across a wide range of characteristics including gene/protein expression and barrier function. Here, we use an isogenic family of fluorescently-labeled iPSC-derived BMEC-like cells (iBMECs) and brain pericyte-like cells (iPCs) within two-dimensional confluent monolayers (2D) and three-dimensional (3D) tissue-engineered microvessels to explore how 3D microenvironment regulates gene expression and function of the in vitro BBB. We show that 3D microenvironment (shear stress, cell-ECM interactions, and cylindrical geometry) increases BBB phenotype and endothelial identity, and alters angiogenic and cytokine responses in synergy with pericyte co-culture. Tissue-engineered microvessels incorporating junction-labeled iBMECs enable study of the real-time dynamics of tight junctions during homeostasis and in response to physical and chemical perturbations.


Assuntos
Barreira Hematoencefálica , Células-Tronco Pluripotentes Induzidas , Barreira Hematoencefálica/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Endoteliais/metabolismo , Junções Íntimas , Diferenciação Celular/fisiologia , Microvasos/metabolismo , Encéfalo/irrigação sanguínea , Expressão Gênica , Células Cultivadas
10.
Adv Sci (Weinh) ; 9(35): e2204395, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36156464

RESUMO

Lyme disease is a tick-borne disease prevalent in North America, Europe, and Asia. Despite the accumulated knowledge from epidemiological, in vitro, and in animal studies, the understanding of dissemination of vector-borne pathogens, such as Borrelia burgdorferi (Bb), remains incomplete with several important knowledge gaps, especially related to invasion and intravasation into circulation. To elucidate the mechanistic details of these processes a tissue-engineered human dermal microvessel model is developed. Fluorescently labeled Bb are injected into the extracellular matrix (ECM) to mimic tick inoculation. High resolution, confocal imaging is performed to visualize the sub-acute phase of infection. From analysis of migration paths no evidence to support adhesin-mediated interactions between Bb and ECM components is found, suggesting that collagen fibers serve as inert obstacles to migration. Intravasation occurs at cell-cell junctions and is relatively fast, consistent with Bb swimming in ECM. In addition, it is found that Bb alone can induce endothelium activation, resulting in increased immune cell adhesion but no changes in global or local permeability. Together these results provide new insight into the minimum requirements for Bb dissemination and highlight how tissue-engineered models are complementary to animal models in visualizing dynamic processes associated with vector-borne pathogens.


Assuntos
Borrelia burgdorferi , Doença de Lyme , Animais , Humanos , Doença de Lyme/microbiologia , Modelos Animais , Microvasos , Pele
11.
Fluids Barriers CNS ; 19(1): 54, 2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35773691

RESUMO

Huntington's disease (HD) is an inherited neurodegenerative disease caused by expansion of cytosine-adenine-guanine (CAG) repeats in the huntingtin gene, which leads to neuronal loss and decline in cognitive and motor function. Increasing evidence suggests that blood-brain barrier (BBB) dysfunction may contribute to progression of the disease. Studies in animal models, in vitro models, and post-mortem tissue find that disease progression is associated with increased microvascular density, altered cerebral blood flow, and loss of paracellular and transcellular barrier function. Here, we report on changes in BBB phenotype due to expansion of CAG repeats using an isogenic pair of induced pluripotent stem cells (iPSCs) differentiated into brain microvascular endothelial-like cells (iBMECs). We show that CAG expansion associated with juvenile HD alters the trajectory of iBMEC differentiation, producing cells with ~ two-fold lower percentage of adherent endothelial cells. CAG expansion is associated with diminished transendothelial electrical resistance and reduced tight junction protein expression, but no significant changes in paracellular permeability. While mutant huntingtin protein (mHTT) aggregates were not observed in HD iBMECs, widespread transcriptional dysregulation was observed in iBMECs compared to iPSCs. In addition, CAG expansion in iBMECs results in distinct responses to pathological and therapeutic perturbations including angiogenic factors, oxidative stress, and osmotic stress. In a tissue-engineered BBB model, iBMECs show subtle changes in phenotype, including differences in cell turnover and immune cell adhesion. Our results further support that CAG expansion in BMECs contributes to BBB dysfunction during HD.


Assuntos
Doença de Huntington , Células-Tronco Pluripotentes Induzidas , Doenças Neurodegenerativas , Animais , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Doença de Huntington/metabolismo , Células-Tronco Pluripotentes Induzidas/fisiologia , Doenças Neurodegenerativas/metabolismo
12.
Fluids Barriers CNS ; 19(1): 33, 2022 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-35551622

RESUMO

Oxidative stress is a shared pathology of neurodegenerative disease and brain injuries, and is derived from perturbations to normal cell processes by aging or environmental factors such as UV exposure and air pollution. As oxidative cues are often present in systemic circulation, the blood-brain barrier (BBB) plays a key role in mediating the effect of these cues on brain dysfunction. Therefore, oxidative damage and disruption of the BBB is an emergent focus of neurodegenerative disease etiology and progression. We assessed barrier dysfunction in response to chronic and acute oxidative stress in 2D and 3D in vitro models of the BBB with human iPSC-derived brain microvascular endothelial-like cells (iBMECs). We first established doses of hydrogen peroxide to induce chronic damage (modeling aging and neurodegenerative disease) and acute damage (modeling the response to traumatic brain injury) by assessing barrier function via transendothelial electrical resistance in 2D iBMEC monolayers and permeability and monolayer integrity in 3D tissue-engineered iBMEC microvessels. Following application of these chronic and acute doses in our in vitro models, we found local, discrete structural changes were the most prevalent responses (rather than global barrier loss). Additionally, we validated unique functional changes in response to oxidative stress, including dysfunctional cell turnover dynamics and immune cell adhesion that were consistent with changes in gene expression.


Assuntos
Barreira Hematoencefálica , Doenças Neurodegenerativas , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Humanos , Microvasos/metabolismo , Doenças Neurodegenerativas/metabolismo , Estresse Oxidativo
13.
Neurobiol Dis ; 169: 105719, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35398340

RESUMO

Oxidative stress is caused by an imbalance between the generation and detoxification of reactive oxygen and nitrogen species (ROS/RNS). This imbalance plays an important role in brain aging and age-related neurodegenerative diseases. In the context of Parkinson's disease (PD), the sensitivity of dopaminergic neurons in the substantia nigra pars compacta to oxidative stress is considered a key factor of PD pathogenesis. Here we study the effect of different oxidative stress-inducing compounds (6-OHDA, MPTP or MPP+) on the population of dopaminergic neurons in an iPSC-derived human brain 3D model (aka BrainSpheres). Treatment with 6-OHDA, MPTP or MPP+ at 4 weeks of differentiation disrupted the dopaminergic neuronal phenotype in BrainSpheres at (50, 5000, 1000 µM respectively). 6-OHDA increased ROS production and decreased mitochondrial function most efficiently. It further induced the greatest changes in gene expression and metabolites related to oxidative stress and mitochondrial dysfunction. Co-culturing BrainSpheres with an endothelial barrier using a transwell system allowed the assessment of differential penetration capacities of the tested compounds and the damage they caused in the dopaminergic neurons within the BrainSpheres In conclusion, treatment with compounds known to induce PD-like phenotypes in vivo caused molecular deficits and loss of dopaminergic neurons in the BrainSphere model. This approach therefore recapitulates common animal models of neurodegenerative processes in PD at similarly high doses. The relevance as tool for drug discovery is discussed.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Substância Negra/metabolismo
14.
FASEB J ; 36(5): e22331, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35476363

RESUMO

The blood-brain barrier (BBB) regulates molecular and cellular entry from the cerebrovasculature into the surrounding brain parenchyma. Many diseases of the brain are associated with dysfunction of the BBB, where hypoxia is a common stressor. However, the contribution of hypoxia to BBB dysfunction is challenging to study due to the complexity of the brain microenvironment. In this study, we used a BBB model with brain microvascular endothelial cells and pericytes differentiated from iPSCs to investigate the effect of hypoxia on barrier function. We found that hypoxia-induced barrier dysfunction is dependent upon increased actomyosin contractility and is associated with increased fibronectin fibrillogenesis. We propose a role for actomyosin contractility in mediating hypoxia-induced barrier dysfunction through modulation of junctional claudin-5. Our findings suggest pericytes may protect brain microvascular endothelial cells from hypoxic stresses and that pericyte-derived factors could be candidates for treatment of pathological barrier-forming tissues.


Assuntos
Actomiosina , Barreira Hematoencefálica , Claudina-5 , Células Endoteliais , Pericitos , Actomiosina/metabolismo , Barreira Hematoencefálica/metabolismo , Hipóxia Celular/efeitos da radiação , Claudina-5/metabolismo , Meios de Cultivo Condicionados , Células Endoteliais/metabolismo , Humanos , Pericitos/metabolismo
15.
JCI Insight ; 7(9)2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35349483

RESUMO

BackgroundSome clinical features of severe COVID-19 represent blood vessel damage induced by activation of host immune responses initiated by the coronavirus SARS-CoV-2. We hypothesized autoantibodies against angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor expressed on vascular endothelium, are generated during COVID-19 and are of mechanistic importance.MethodsIn an opportunity sample of 118 COVID-19 inpatients, autoantibodies recognizing ACE2 were detected by ELISA. Binding properties of anti-ACE2 IgM were analyzed via biolayer interferometry. Effects of anti-ACE2 IgM on complement activation and endothelial function were demonstrated in a tissue-engineered pulmonary microvessel model.ResultsAnti-ACE2 IgM (not IgG) autoantibodies were associated with severe COVID-19 and found in 18/66 (27.2%) patients with severe disease compared with 2/52 (3.8%) of patients with moderate disease (OR 9.38, 95% CI 2.38-42.0; P = 0.0009). Anti-ACE2 IgM autoantibodies were rare (2/50) in non-COVID-19 ventilated patients with acute respiratory distress syndrome. Unexpectedly, ACE2-reactive IgM autoantibodies in COVID-19 did not undergo class-switching to IgG and had apparent KD values of 5.6-21.7 nM, indicating they are T cell independent. Anti-ACE2 IgMs activated complement and initiated complement-binding and functional changes in endothelial cells in microvessels, suggesting they contribute to the angiocentric pathology of COVID-19.ConclusionWe identify anti-ACE2 IgM as a mechanism-based biomarker strongly associated with severe clinical outcomes in SARS-CoV-2 infection, which has therapeutic implications.FUNDINGBill & Melinda Gates Foundation, Gates Philanthropy Partners, Donald B. and Dorothy L. Stabler Foundation, and Jerome L. Greene Foundation; NIH R01 AR073208, R01 AR069569, Institutional Research and Academic Career Development Award (5K12GM123914-03), National Heart, Lung, and Blood Institute R21HL145216, and Division of Intramural Research, National Institute of Allergy and Infectious Diseases; National Science Foundation Graduate Research Fellowship (DGE1746891).


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Autoanticorpos , Células Endoteliais , Humanos , Imunoglobulina M , SARS-CoV-2
16.
ACS Omega ; 7(1): 1100-1112, 2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35036773

RESUMO

The blood-brain barrier remains a major roadblock to the delivery of drugs to the brain. While in vitro and in vivo measurements of permeability are widely used to predict brain penetration, very little is known about the mechanisms of passive transport. Detailed insight into interactions between solutes and cell membranes could provide new insight into drug design and screening. Here, we perform unbiased atomistic MD simulations to visualize translocation of a library of 24 solutes across a lipid bilayer representative of brain microvascular endothelial cells. A temperature bias is used to achieve steady state of all solutes, including those with low permeability. Based on free-energy surface profiles, we show that the solutes can be classified into three groups that describe distinct mechanisms of transport across the bilayer. Simulations down to 310 K for solutes with fast permeability were used to justify the extrapolation of values at 310 K from higher temperatures. Comparison of permeabilities at 310 K to experimental values obtained from in vitro transwell measurements and in situ brain perfusion revealed that permeabilities obtained from simulations vary from close to the experimental values to more than 3 orders of magnitude faster. The magnitude of the difference was dependent on the group defined by free-energy surface profiles. Overall, these results show that MD simulations can provide new insight into the mechanistic details of brain penetration and provide a new approach for drug discovery.

17.
Fluids Barriers CNS ; 18(1): 56, 2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34876171

RESUMO

With the limitations associated with post-mortem tissue and animal models, In vitro BBB models enable precise control of independent variables and microenvironmental cues, and hence play an important role in studying the BBB. Advances in stem cell technology and tissue engineering provide the tools to create next-generation in vitro BBB models with spatial organization of different cell types in 3D microenvironments that more closely match the human brain. These models will be capable of assessing the physiological and pathological responses to different perturbations relevant to health and disease. Here, we review the factors that determine the accuracy of in vitro BBB models, and describe how these factors will guide the development of next-generation models. Improving the accuracy of cell sources and microenvironmental cues will enable in vitro BBB models with improved accuracy and specificity to study processes and phenomena associated with zonation, brain region, age, sex, ethnicity, and disease state.


Assuntos
Barreira Hematoencefálica , Microambiente Celular , Células Endoteliais , Modelos Biológicos , Células-Tronco Pluripotentes , Animais , Benchmarking , Humanos
18.
Biomaterials ; 275: 120942, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34147718

RESUMO

The blood-brain barrier (BBB) tightly controls entry of molecules and cells into the brain, restricting the delivery of therapeutics. Blood-brain barrier opening (BBBO) utilizes reversible disruption of cell-cell junctions between brain microvascular endothelial cells to enable transient entry into the brain. Here, we demonstrate that melittin, a membrane active peptide present in bee venom, supports transient BBBO. From endothelial and neuronal viability studies, we first identify the accessible concentration range for BBBO. We then use a tissue-engineered model of the human BBB to optimize dosing and elucidate the mechanism of opening. Melittin and other membrane active variants transiently increase paracellular permeability via disruption of cell-cell junctions that result in transient focal leaks. To validate the results from the tissue-engineered model, we then demonstrate that transient BBBO can be reproduced in a mouse model. We identify a minimum clinically effective intra-arterial dose of 3 µM min melittin, which is reversible within one day and neurologically safe. Melittin-induced BBBO represents a novel technology for delivery of therapeutics into the brain.


Assuntos
Barreira Hematoencefálica , Meliteno , Animais , Transporte Biológico , Encéfalo , Células Endoteliais , Camundongos
19.
ACS Sens ; 5(12): 3821-3826, 2020 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-33263987

RESUMO

Individualized measurement of sweat loss under heat stress is important in assessing physical performance and preventing heat-related illness for athletes or individuals working in extreme environments. The objective of this work was to develop a low-cost and easy-to-fabricate wearable sensor that enables accurate real-time measurement of sweat rate. A capacitive-type sensor was fabricated from two conducting parallel plates, plastic insulating layers, and a central microfluidic channel formed by laser cutting a plastic film. The device has no microfabricated electrodes and is assembled using adhesive tape. Sensor accuracy was validated at different flow rates and confirmed using an equivalent circuit model of the device. On-body measurements demonstrate the feasibility of real-time measurements and show good agreement with values determined from a conventional sweat collection device.


Assuntos
Suor , Dispositivos Eletrônicos Vestíveis , Eletrodos , Humanos
20.
Mol Pharm ; 17(9): 3425-3434, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32787285

RESUMO

Brain microvascular endothelial cells derived from induced pluripotent stem cells (dhBMECs) are a scalable and reproducible resource for studies of the human blood-brain barrier, including mechanisms and strategies for drug delivery. Confluent monolayers of dhBMECs recapitulate key in vivo functions including tight junctions to limit paracellular permeability and efflux and nutrient transport to regulate transcellular permeability. Techniques for cryopreservation of dhBMECs have been reported; however, functional validation studies after long-term cryopreservation have not been extensively performed. Here, we characterize dhBMECs after 1 year of cryopreservation using selective purification on extracellular matrix-treated surfaces and ROCK inhibition. One-year cryopreserved dhBMECs maintain functionality of tight junctions, efflux pumps, and nutrient transporters with stable protein localization and gene expression. Cryopreservation is associated with a decrease in the yield of adherent cells and unique responses to cell stress, resulting in altered paracellular permeability of Lucifer yellow. Additionally, cryopreserved dhBMECs reliably form functional three-dimensional microvessels independent of cryopreservation length, with permeabilities lower than non-cryopreserved two-dimensional models. Long-term cryopreservation of dhBMECs offers key advantages including increased scalability, reduced batch-to-batch effects, the ability to conduct well-controlled follow up studies, and support of multisite collaboration from the same cell stock, all while maintaining phenotype for screening pharmaceutical agents.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/fisiologia , Células Endoteliais/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Microvasos/fisiologia , Transporte Biológico/fisiologia , Permeabilidade Capilar/fisiologia , Células Cultivadas , Criopreservação/métodos , Matriz Extracelular/fisiologia , Expressão Gênica/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Junções Íntimas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...