Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neurodev Disord ; 16(1): 27, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783199

ABSTRACT

BACKGROUND: Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene and dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB). METHODS: We generated TSC disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies. RESULTS: Using microphysiological systems, we demonstrate that a BBB generated from TSC2 heterozygous mutant cells shows increased permeability. This can be rescued by wild type astrocytes or by treatment with rapamycin, an mTOR kinase inhibitor. CONCLUSION: Our results demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of cell lineages contributing to TSC pathogenesis and informs future therapeutics.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Tuberous Sclerosis Complex 2 Protein , Tuberous Sclerosis , Tuberous Sclerosis/physiopathology , Tuberous Sclerosis/genetics , Humans , Blood-Brain Barrier/physiopathology , Tuberous Sclerosis Complex 2 Protein/genetics , Sirolimus/pharmacology , Astrocytes/metabolism
2.
Lab Chip ; 24(6): 1794-1807, 2024 03 12.
Article in English | MEDLINE | ID: mdl-38362777

ABSTRACT

Human microphysiological systems, such as organs on chips, are an emerging technology for modeling human physiology in a preclinical setting to understand the mechanism of action of drugs, to evaluate the efficacy of treatment options for human disease and impairment, and to assess drug toxicity. By using human cells co-cultured in three-dimensional constructs, organ chips can provide greater fidelity to the human cellular condition than their two-dimensional predecessors. However, with the rise of SARS-CoV-2 and the global COVID-19 pandemic, it became clear that many microphysiological systems were not compatible with or optimized for studies of infectious disease and operation in a Biosafety Level 3 (BSL-3) environment. Given that one of the early sites of SARS-CoV-2 infection is the airway, we created a human airway organ chip that could operate in a BSL-3 space with high throughput and minimal manipulation, while retaining the necessary physical and physiological components to recapitulate tissue response to infectious agents and the immune response to infection.


Subject(s)
COVID-19 , Humans , SARS-CoV-2 , Viral Load , Pandemics , Immunohistochemistry , Cytokines , Lab-On-A-Chip Devices
3.
bioRxiv ; 2023 Dec 16.
Article in English | MEDLINE | ID: mdl-38168450

ABSTRACT

Tuberous sclerosis complex (TSC) is a multi-system genetic disease that causes benign tumors in the brain and other vital organs. The most debilitating symptoms result from involvement of the central nervous system and lead to a multitude of severe symptoms including seizures, intellectual disability, autism, and behavioral problems. TSC is caused by heterozygous mutations of either the TSC1 or TSC2 gene. Dysregulation of mTOR kinase with its multifaceted downstream signaling alterations is central to disease pathogenesis. Although the neurological sequelae of the disease are well established, little is known about how these mutations might affect cellular components and the function of the blood-brain barrier (BBB). We generated disease-specific cell models of the BBB by leveraging human induced pluripotent stem cell and microfluidic cell culture technologies. Using these microphysiological systems, we demonstrate that the BBB generated from TSC2 heterozygous mutant cells shows increased permeability which can be rescued by wild type astrocytes and with treatment with rapamycin, an mTOR kinase inhibitor. Our results further demonstrate the utility of microphysiological systems to study human neurological disorders and advance our knowledge of the cell lineages contributing to TSC pathogenesis.

4.
Fluids Barriers CNS ; 17(1): 38, 2020 Jun 03.
Article in English | MEDLINE | ID: mdl-32493346

ABSTRACT

BACKGROUND: The United States faces a national crisis involving opioid medications, where currently more than 130 people die every day. To combat this epidemic, a better understanding is needed of how opioids penetrate into the central nervous system (CNS) to facilitate pain relief and, potentially, result in addiction and/or misuse. Animal models, however, are a poor predictor of blood-brain barrier (BBB) transport and CNS drug penetration in humans, and many traditional 2D cell culture models of the BBB and neurovascular unit have inadequate barrier function and weak or inappropriate efflux transporter expression. Here, we sought to better understand opioid transport mechanisms using a simplified microfluidic neurovascular unit (NVU) model consisting of human brain microvascular endothelial cells (BMECs) co-cultured with astrocytes. METHODS: Human primary and induced pluripotent stem cell (iPSC)-derived BMECs were incorporated into a microfluidic NVU model with several technical improvements over our previous design. Passive barrier function was assessed by permeability of fluorescent dextrans with varying sizes, and P-glycoprotein function was assessed by rhodamine permeability in the presence or absence of inhibitors; quantification was performed with a fluorescent plate reader. Loperamide, morphine, and oxycodone permeability was assessed in the presence or absence of P-glycoprotein inhibitors and cortisol; quantification was performed with mass spectrometry. RESULTS: We first report technical and methodological optimizations to our previously described microfluidic model using primary human BMECs, which results in accelerated barrier formation, decreased variability, and reduced passive permeability relative to Transwell models. We then demonstrate proper transport and efflux of loperamide, morphine, and oxycodone in the microfluidic NVU containing BMECs derived from human iPSCs. We further demonstrate that cortisol can alter permeability of loperamide and morphine in a divergent manner. CONCLUSIONS: We reveal a novel role for the stress hormone cortisol in modulating the transport of opioids across the BBB, which could contribute to their abuse or overdose. Our updated BBB model represents a powerful tool available to researchers, clinicians, and drug manufacturers for understanding the mechanisms by which opioids access the CNS.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Astrocytes/physiology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Hydrocortisone/metabolism , Induced Pluripotent Stem Cells/physiology , Models, Neurological , Astrocytes/drug effects , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Microvessels/cytology
5.
Stem Cell Reports ; 12(3): 474-487, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30773484

ABSTRACT

There is a profound need for functional, biomimetic in vitro tissue constructs of the human blood-brain barrier and neurovascular unit (NVU) to model diseases and identify therapeutic interventions. Here, we show that induced pluripotent stem cell (iPSC)-derived human brain microvascular endothelial cells (BMECs) exhibit robust barrier functionality when cultured in 3D channels within gelatin hydrogels. We determined that BMECs cultured in 3D under perfusion conditions were 10-100 times less permeable to sodium fluorescein, 3 kDa dextran, and albumin relative to human umbilical vein endothelial cell and human dermal microvascular endothelial cell controls, and the BMECs maintained barrier function for up to 21 days. Analysis of cell-cell junctions revealed expression patterns supporting barrier formation. Finally, efflux transporter activity was maintained over 3 weeks of perfused culture. Taken together, this work lays the foundation for development of a representative 3D in vitro model of the human NVU constructed from iPSCs.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Endothelial Cells/drug effects , Endothelium/drug effects , Hydrogels/pharmacology , Induced Pluripotent Stem Cells/drug effects , Albumins/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Cells, Cultured , Dextrans/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Fluorescein/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Microvessels/drug effects , Microvessels/metabolism
6.
SLAS Technol ; 23(6): 592-598, 2018 12.
Article in English | MEDLINE | ID: mdl-29787331

ABSTRACT

The fabrication of engineered vascularized tissues and organs requiring sustained, controlled perfusion has been facilitated by the development of several pump systems. Currently, researchers in the field of tissue engineering require the use of pump systems that are in general large, expensive, and generically designed. Overall, these pumps often fail to meet the unique demands of perfusing clinically useful tissue constructs. Here, we describe a pumping platform that overcomes these limitations and enables scalable perfusion of large, three-dimensional hydrogels. We demonstrate the ability to perfuse multiple separate channels inside hydrogel slabs using a preprogrammed schedule that dictates pumping speed and time. The use of this pump system to perfuse channels in large-scale engineered tissue scaffolds sustained cell viability over several weeks.


Subject(s)
Hydrogels , Perfusion/methods , Tissue Culture Techniques/methods , Tissue Engineering/methods , Costs and Cost Analysis , Perfusion/economics , Perfusion/instrumentation , Tissue Culture Techniques/economics , Tissue Culture Techniques/instrumentation , Tissue Engineering/economics , Tissue Engineering/instrumentation
7.
Article in English | MEDLINE | ID: mdl-29441348

ABSTRACT

The neurovascular unit (NVU) is composed of neurons, astrocytes, pericytes, and endothelial cells that form the blood-brain barrier (BBB). The NVU regulates material exchange between the bloodstream and the brain parenchyma, and its dysfunction is a primary or secondary cause of many cerebrovascular and neurodegenerative disorders. As such, there are substantial research thrusts in academia and industry toward building NVU models that mimic endogenous organization and function, which could be used to better understand disease mechanisms and assess drug efficacy. Human pluripotent stem cells, which can self-renew indefinitely and differentiate to almost any cell type in the body, are attractive for these models because they can provide a limitless source of individual cells from the NVU. In addition, human-induced pluripotent stem cells (iPSCs) offer the opportunity to build NVU models with an explicit genetic background and in the context of disease susceptibility. Herein, we review how iPSCs are being used to model neurovascular and neurodegenerative diseases, with particular focus on contributions of the BBB, and discuss existing technologies and emerging opportunities to merge these iPSC progenies with biomaterials platforms to create complex NVU systems that recreate the in vivo microenvironment.

8.
Biomicrofluidics ; 9(3): 036501, 2015 May.
Article in English | MEDLINE | ID: mdl-26045731

ABSTRACT

Biomimetic scaffolds approaching physiological scale, whose size and large cellular load far exceed the limits of diffusion, require incorporation of a fluidic means to achieve adequate nutrient/metabolite exchange. This need has driven the extension of microfluidic technologies into the area of biomaterials. While construction of perfusable scaffolds is essentially a problem of microfluidic device fabrication, functional implementation of free-standing, thick-tissue constructs depends upon successful integration of external pumping mechanisms through optimized connective assemblies. However, a critical analysis to identify optimal materials/assembly components for hydrogel substrates has received little focus to date. This investigation addresses this issue directly by evaluating the efficacy of a range of adhesive and mechanical fluidic connection methods to gelatin hydrogel constructs based upon both mechanical property analysis and cell compatibility. Results identify a novel bioadhesive, comprised of two enzymatically modified gelatin compounds, for connecting tubing to hydrogel constructs that is both structurally robust and non-cytotoxic. Furthermore, outcomes from this study provide clear evidence that fluidic interconnect success varies with substrate composition (specifically hydrogel versus polydimethylsiloxane), highlighting not only the importance of selecting the appropriately tailored components for fluidic hydrogel systems but also that of encouraging ongoing, targeted exploration of this issue. The optimization of such interconnect systems will ultimately promote exciting scientific and therapeutic developments provided by microfluidic, cell-laden scaffolds.

9.
Biomicrofluidics ; 5(2): 24106, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21673844

ABSTRACT

We discuss the ability to perform fluorescent immunocytochemistry, following cell fixation, using a microfluidic array of primary, nonadherent, single CD34+ stem cells. The technique requires small cell samples and proceeds with no cell loss, making it well-suited to monitoring these rare patient-derived cells. The chip allows us to correlate live cell dynamics across arrays of individual cells with post-translational modifications of intracellular proteins, following their exposure to drug treatments. Results also show that due to the microfluidic environment, the time scale of cell fixation was significantly reduced compared to conventional methods, leading to greater confidence in the status of the protein modifications studied.

10.
Chem Commun (Camb) ; 46(42): 7921-3, 2010 Nov 14.
Article in English | MEDLINE | ID: mdl-20859575

ABSTRACT

We report for the first time the time-resolved mapping of intracellular nanoparticle labels from within living cells retained in a microstructured trap using Raman spectroscopy. The methods employed here also demonstrate the ability to rapidly discriminate between cell populations containing different SERS labels.


Subject(s)
Microfluidics/instrumentation , Nanoparticles , Animals , CHO Cells , Cricetinae , Cricetulus , Spectrum Analysis, Raman
11.
Exp Biol Med (Maywood) ; 235(6): 777-83, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20511682

ABSTRACT

Microfluidic devices are well-suited for the study of metabolism and paracrine and autocrine signaling because they allow steady or intermittent perfusion of biological cells at cell densities that approach those in living tissue. They also enable the study of small populations of rare cells. However, it can be difficult to introduce the cells into a microfluidic device to achieve and control such densities without damaging or clumping the cells. We describe simple procedures that address the problem of efficient introduction of cells and cell culture media into microfluidic devices using small bore polyetheretherketone (PEEK) tubing and Hamilton gastight syringes. Suspension or adherent cells grown in cell culture flasks are centrifuged and extracted directly from the centrifuge pellet into the end of the PEEK tubing by aspiration. The tube end is then coupled to prepunched channels in the polydimethylsiloxane microfluidic device by friction fitting. Controlled depression of the syringe plunger expels the cells into the microfluidic device only seconds following aspiration. The gastight syringes and PEEK tubing with PEEK fittings provide a non-compliant source of pressure and suction with a rapid response time that is well suited for short-term intramicrofluidic cellular studies. The benefits of this method are its simplicity, modest expense, the short preparation time required for loading appropriate numbers of cells and the applicability of the technique to small quantities of rare or expensive cells. This should in turn lead to new applications of microfluidic devices to biology and medicine.


Subject(s)
Microfluidics , Cell Culture Techniques/methods , Cell Line , Cells, Cultured , Centrifugation
12.
Lab Chip ; 9(18): 2659-64, 2009 Sep 21.
Article in English | MEDLINE | ID: mdl-19704981

ABSTRACT

Stem cells hold great promise as a means of treating otherwise incurable, degenerative diseases due to their ability both to self-renew and differentiate. However, stem cell damage can also play a role in the disease with the formation of solid tumors and leukaemias such as chronic myeloid leukaemia (CML), a hematopoietic stem cell (HSC) disorder. Despite recent medical advances, CML remains incurable by drug therapy. Understanding the mechanisms which govern chemoresistance of individual stem cell leukaemias may therefore require analysis at the single cell level. This task is not trivial using current technologies given that isolating HSCs is difficult, expensive, and inefficient due to low cell yield from patients. In addition, hematopoietic cells are largely non-adherent and thus difficult to study over time using conventional cell culture techniques. Hence, there is a need for new microfluidic platforms that allow the functional interrogation of hundreds of non-adherent single cells in parallel. We demonstrate the ability to perform assays, normally performed on the macroscopic scale, within the microfluidic platform using minimal reagents and low numbers of primary cells. We investigated normal and CML stem cell responses to the tyrosine kinase inhibitor, dasatinib, a drug approved for the treatment of CML. Dynamic, on-chip three-color cell viability assays revealed that differences in the responses of normal and CML stem/progenitor cells to dasatinib were observed even in the early phases of exposure, during which time normal cells exhibit a significantly elevated cell death rate, as compared to both controls and CML cells. Further studies show that dasatinib does, however, markedly reduce CML stem/progenitor cell migration in situ.


Subject(s)
Hematopoietic Stem Cells/physiology , Microfluidics/methods , Annexin A5/metabolism , Apoptosis/physiology , Cell Division/physiology , Cell Survival , Cells, Cultured , Dasatinib , Humans , Kinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Microscopy, Fluorescence , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Signal Transduction/physiology , Thiazoles/pharmacology , Thiazoles/therapeutic use
13.
Mol Imaging ; 6(5): 331-9, 2007.
Article in English | MEDLINE | ID: mdl-18092518

ABSTRACT

Vascular endothelial growth factor (VEGF) is a major inducer of angiogenesis. We generated a transgenic reporter mouse, VEGF-GL, in which an enhanced green fluorescent protein-luciferase fusion protein is expressed under the control of a human VEGF-A promoter. The VEGF-GL mouse exhibited intense bioluminescence throughout the body at 1 week of age. The signals rapidly declined to a relatively low level as the mice grew. The adult VEGF-GL mouse showed restricted bioluminescence to the areas undergoing wound healing. In contrast, the VEGF-GL mice, which were crossed with mouse mammary tumor virus-polyoma virus middle T antigen transgenic mammary tumor mice, exhibited prominent bioluminescence in the tumors, correlating with VEGF transcription. Tumor bioluminescence was observed in the bigenic mice as early as 8 weeks, before tumors were palpable, and the signals increased with tumor growth. In conclusion, the VEGF-GL mouse permits longitudinal and quantitative assessment of VEGF promoter activity in vivo. The model should facilitate understanding of the molecular controls and pathways that regulate VEGF transcription in vivo.


Subject(s)
Luminescent Measurements/methods , Mammary Neoplasms, Animal/pathology , Promoter Regions, Genetic/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Blotting, Western , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoprecipitation , In Situ Hybridization , Luciferases/genetics , Luciferases/metabolism , Male , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Transgenic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
14.
IEEE Trans Nanobioscience ; 5(4): 268-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17181026

ABSTRACT

Antibody-conjugated quantum dots (QDs) have been used to map the expression dynamics of the cytokine receptor interleukin-2 receptor-alpha (IL-2Ralpha) following Jurkat T cell activation. Maximal receptor expression was observed 48 h after activation, followed by a sharp decrease consistent with IL-2R internalization subsequent to IL-2 engagement. Verification of T cell activation and specificity of QD labeling were demonstrated using fluorescence microscopy, ELISA, and FACS analyses. These antibody conjugates provide a versatile means to rapidly determine cell state and interrogate membrane associated proteins involved in cell signaling pathways. Ultimately, incorporation with a microfluidic platform capable of simultaneously monitoring several cell signaling pathways will aid in toxin detection and discrimination.


Subject(s)
Cytokines/immunology , Lymphocyte Activation/immunology , Microscopy, Fluorescence/methods , Molecular Probe Techniques , Quantum Dots , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Humans , Jurkat Cells , Receptors, Cytokine/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...