1
|
Shamul JG, Wang Z, Gong H, Ou W, White AM, Moniz-Garcia DP, Gu S, Clyne AM, Quiñones-Hinojosa A, He X. Meta-analysis of the make-up and properties of in vitro models of the healthy and diseased blood-brain barrier. Nat Biomed Eng 2024:10.1038/s41551-024-01250-2. [PMID: 39304761 DOI: 10.1038/s41551-024-01250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/08/2024] [Indexed: 09/22/2024]
Abstract
In vitro models of the human blood-brain barrier (BBB) are increasingly used to develop therapeutics that can cross the BBB for treating diseases of the central nervous system. Here we report a meta-analysis of the make-up and properties of transwell and microfluidic models of the healthy BBB and of BBBs in glioblastoma, Alzheimer's disease, Parkinson's disease and inflammatory diseases. We found that the type of model, the culture method (static or dynamic), the cell types and cell ratios, and the biomaterials employed as extracellular matrix are all crucial to recapitulate the low permeability and high expression of tight-junction proteins of the BBB, and to obtain high trans-endothelial electrical resistance. Specifically, for models of the healthy BBB, the inclusion of endothelial cells and pericytes as well as physiological shear stresses (~10-20 dyne cm-2) are necessary, and when astrocytes are added, astrocytes or pericytes should outnumber endothelial cells. We expect this meta-analysis to facilitate the design of increasingly physiological models of the BBB.
Collapse
Affiliation(s)
- James G Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Zhiyuan Wang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Hyeyeon Gong
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Alisa M White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | | | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA
| | | | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA.
- Brain and Behavior Institute, University of Maryland, College Park, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
2
|
Khalil A, Barras A, Boukherroub R, Tseng CL, Devos D, Burnouf T, Neuhaus W, Szunerits S. Enhancing paracellular and transcellular permeability using nanotechnological approaches for the treatment of brain and retinal diseases. NANOSCALE HORIZONS 2023; 9:14-43. [PMID: 37853828 DOI: 10.1039/d3nh00306j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Paracellular permeability across epithelial and endothelial cells is, in large part, regulated by apical intercellular junctions also referred to as tight junctions (TJs). These junctions contribute to the spatial definition of different tissue compartments within organisms, separating them from the outside world as well as from inner compartments, with their primary physiological role of maintaining tissue homeostasis. TJs restrict the free, passive diffusion of ions and hydrophilic small molecules through paracellular clefts and are important for appropriate cell polarization and transporter protein localisation, supporting the controlled transcellular diffusion of smaller and larger hydrophilic as well as hydrophobic substances. This traditional diffusion barrier concept of TJs has been challenged lately, owing to a better understanding of the components that are associated with TJs. It is now well-established that mutations in TJ proteins are associated with a range of human diseases and that a change in the membrane fluidity of neighbouring cells can open possibilities for therapeutics to cross intercellular junctions. Nanotechnological approaches, exploiting ultrasound or hyperosmotic agents and permeation enhancers, are the paradigm for achieving enhanced paracellular diffusion. The other widely used transport route of drugs is via transcellular transport, allowing the passage of a variety of pro-drugs and nanoparticle-encapsulated drugs via different mechanisms based on receptors and others. For a long time, there was an expectation that lipidic nanocarriers and polymeric nanostructures could revolutionize the field for the delivery of RNA and protein-based therapeutics across different biological barriers equipped with TJs (e.g., blood-brain barrier (BBB), retina-blood barrier (RBB), corneal TJs, etc.). However, only a limited increase in therapeutic efficiency has been reported for most systems until now. The purpose of this review is to explore the reasons behind the current failures and to examine the emergence of synthetic and cell-derived nanomaterials and nanotechnological approaches as potential game-changers in enhancing drug delivery to target locations both at and across TJs using innovative concepts. Specifically, we will focus on recent advancements in various nanotechnological strategies enabling the bypassing or temporally opening of TJs to the brain and to the retina, and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Asmaa Khalil
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Alexandre Barras
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Ching-Li Tseng
- Taipei Medical University, Graduate Institute of Biomedical Materials and Tissue Engineering (GIBMTE), New Taipei City 235603, Taiwan
- Taipei Medical University, International PhD Program in Biomedical Engineering (IPBME), New Taipei City 235603, Taiwan
| | - David Devos
- University Lille, CHU-Lille, Inserm, U1172, Lille Neuroscience & Cognition, LICEND, Lille, France
| | - Thierry Burnouf
- Taipei Medical University, Graduate Institute of Biomedical Materials and Tissue Engineering (GIBMTE), New Taipei City 235603, Taiwan
- Taipei Medical University, International PhD Program in Biomedical Engineering (IPBME), New Taipei City 235603, Taiwan
| | - Winfried Neuhaus
- AIT - Austrian Institute of Technology GmbH, Center Health and Bioresources, Competence Unit Molecular Diagnostics, 1210 Vienna, Austria
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
| | - Sabine Szunerits
- Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| |
Collapse
|
3
|
Park W, Lee JS, Gao G, Kim BS, Cho DW. 3D bioprinted multilayered cerebrovascular conduits to study cancer extravasation mechanism related with vascular geometry. Nat Commun 2023; 14:7696. [PMID: 38001146 PMCID: PMC10673893 DOI: 10.1038/s41467-023-43586-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Cerebral vessels are composed of highly complex structures that facilitate blood perfusion necessary for meeting the high energy demands of the brain. Their geometrical complexities alter the biophysical behavior of circulating tumor cells in the brain, thereby influencing brain metastasis. However, recapitulation of the native cerebrovascular microenvironment that shows continuities between vascular geometry and metastatic cancer development has not been accomplished. Here, we apply an in-bath 3D triaxial bioprinting technique and a brain-specific hybrid bioink containing an ionically crosslinkable hydrogel to generate a mature three-layered cerebrovascular conduit with varying curvatures to investigate the physical and molecular mechanisms of cancer extravasation in vitro. We show that more tumor cells adhere at larger vascular curvature regions, suggesting that prolongation of tumor residence time under low velocity and wall shear stress accelerates the molecular signatures of metastatic potential, including endothelial barrier disruption, epithelial-mesenchymal transition, inflammatory response, and tumorigenesis. These findings provide insights into the underlying mechanisms driving brain metastases and facilitate future advances in pharmaceutical and medical research.
Collapse
Affiliation(s)
- Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea.
- Medical Research Institute, Pusan National University, Yangsan, Republic of Korea.
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea.
| |
Collapse
|
4
|
Jorgensen C, Troendle EP, Ulmschneider JP, Searson PC, Ulmschneider MB. A least-squares-fitting procedure for an efficient preclinical ranking of passive transport across the blood-brain barrier endothelium. J Comput Aided Mol Des 2023; 37:537-549. [PMID: 37573260 PMCID: PMC10505096 DOI: 10.1007/s10822-023-00525-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/24/2023] [Indexed: 08/14/2023]
Abstract
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.
Collapse
Affiliation(s)
- Christian Jorgensen
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Chemistry, Aarhus University, Langelandsgade 140, 8000, Aarhus C, Denmark.
| | | | | | - Peter C Searson
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
5
|
Yan L, Dwiggins CW, Gupta U, Stroka KM. A Rapid-Patterning 3D Vessel-on-Chip for Imaging and Quantitatively Analyzing Cell-Cell Junction Phenotypes. Bioengineering (Basel) 2023; 10:1080. [PMID: 37760182 PMCID: PMC10525190 DOI: 10.3390/bioengineering10091080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the molecular exchanges between the brain and peripheral blood. The permeability of the BBB is primarily regulated by the junction proteins on the brain endothelial cells. In vitro BBB models have shown great potential for the investigation of the mechanisms of physiological function, pathologies, and drug delivery in the brain. However, few studies have demonstrated the ability to monitor and evaluate the barrier integrity by quantitatively analyzing the junction presentation in 3D microvessels. This study aimed to fabricate a simple vessel-on-chip, which allows for a rigorous quantitative investigation of junction presentation in 3D microvessels. To this end, we developed a rapid protocol that creates 3D microvessels with polydimethylsiloxane and microneedles. We established a simple vessel-on-chip model lined with human iPSC-derived brain microvascular endothelial-like cells (iBMEC-like cells). The 3D image of the vessel structure can then be "unwrapped" and converted to 2D images for quantitative analysis of cell-cell junction phenotypes. Our findings revealed that 3D cylindrical structures altered the phenotype of tight junction proteins, along with the morphology of cells. Additionally, the cell-cell junction integrity in our 3D models was disrupted by the tumor necrosis factor α. This work presents a "quick and easy" 3D vessel-on-chip model and analysis pipeline, together allowing for the capability of screening and evaluating the cell-cell junction integrity of endothelial cells under various microenvironment conditions and treatments.
Collapse
Affiliation(s)
- Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
| | - Cole W. Dwiggins
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
| | - Udit Gupta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
- Biophysics Program, University of Maryland, College Park, MD 20742, USA
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Lee EJ, Krassin ZL, Abaci HE, Mahler GJ, Esch MB. Pumped and pumpless microphysiological systems to study (nano)therapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1911. [PMID: 37464464 PMCID: PMC11323280 DOI: 10.1002/wnan.1911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 07/20/2023]
Abstract
Fluidic microphysiological systems (MPS) are microfluidic cell culture devices that are designed to mimic the biochemical and biophysical in vivo microenvironments of human tissues better than conventional petri dishes or well-plates. MPS-grown tissue cultures can be used for probing new drugs for their potential primary and secondary toxicities as well as their efficacy. The systems can also be used for assessing the effects of environmental nanoparticles and nanotheranostics, including their rate of uptake, biodistribution, elimination, and toxicity. Pumpless MPS are a group of MPS that often utilize gravity to recirculate cell culture medium through their microfluidic networks, providing some advantages, but also presenting some challenges. They can be operated with near-physiological amounts of blood surrogate (i.e., cell culture medium) that can recirculate in bidirectional or unidirectional flow patterns depending on the device configuration. Here we discuss recent advances in the design and use of both pumped and pumpless MPS with a focus on where pumpless devices can contribute to realizing the potential future role of MPS in evaluating nanomaterials. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Department of Chemistry and Biochemistry, College of Computer, Mathematical and Natural Sciences, University of Maryland, College Park, Maryland, USA
- Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Zachary L Krassin
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York, USA
| | - Hasan Erbil Abaci
- Department of Dermatology, Columbia University Medical Center, New York, New York, USA
| | - Gretchen J Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, New York, USA
| | - Mandy B Esch
- Microsystems and Nanotechnology Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
7
|
Sekulic M, Abdollahi N, Graf L, Deigendesch N, Puche R, Bodmer D, Petkovic V. Human blood-labyrinth barrier on a chip: a unique in vitro tool for investigation of BLB properties. RSC Adv 2023; 13:25508-25517. [PMID: 37636514 PMCID: PMC10450574 DOI: 10.1039/d3ra04704k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Hearing loss is one of the leading causes of disability worldwide, usually as a result of hair cell damage in the inner ear due to aging, acoustic trauma, or exposure to antibiotics or chemotherapy. No drug therapies can protect or restore hearing and current in vitro and animal models used in drug discovery have a very low success rate, mostly due to major differences in anatomy and accessibility of the inner ear environment between species. The blood-labyrinth barrier (BLB) in the stria vascularis is a highly specialized capillary network that controls exchanges between the blood and interstitial space in the cochlea. The BLB is critical for normal hearing, functioning as a physical, transport, and metabolic barrier. To address its complexity and accessibility, we created the first micro-engineered human model of BLB on a chip using autogenous progenitor cells from adult temporal bones. We successfully isolated the BLB from post-mortem human tissue and established an endothelial cell and pericyte culture system on a BLB chip. Using biocompatible materials, we fabricated sustainable two chamber chips. We validated the size-dependent permeability limits of our BLB model by measuring the permeability to daptomycin (molecular weight 1.6 kDa) and midazolam (molecular weight 325.78 Da). Daptomycin did not pass through the BLB layer, whereas midazolam readily passed through the BLB in our system. Thus, our BLB-chip mimicked the integrity and permeability of human stria vascularis capillaries. This represents a major step towards establishing a reliable model for the development of hearing loss treatments.
Collapse
Affiliation(s)
- Marijana Sekulic
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| | - Narjes Abdollahi
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| | - Lukas Graf
- Clinic for Otolaryngology, Head and Neck Surgery, University Hospital Basel Basel Switzerland
| | | | - Raoul Puche
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| | - Daniel Bodmer
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
- Clinic for Otolaryngology, Head and Neck Surgery, University Hospital Basel Basel Switzerland
| | - Vesna Petkovic
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| |
Collapse
|
8
|
Chavarria D, Abbaspour A, Celestino N, Shah N, Sankar S, Baker AB. A high throughput blood-brain barrier model incorporating shear stress with improved predictive power for drug discovery. BIOMICROFLUIDICS 2023; 17:044105. [PMID: 37614679 PMCID: PMC10444201 DOI: 10.1063/5.0150887] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023]
Abstract
The blood-brain barrier is a key structure regulating the health of the brain and access of drugs and pathogens to neural tissue. Shear stress is a key regulator of the blood-brain barrier; however, the commonly used multi-well vitro models of the blood-brain barrier do not incorporate shear stress. In this work, we designed and validated a high-throughput system for simulating the blood-brain barrier that incorporates physiological flow and incorporates an optimized cellular model of the blood-brain barrier. This system can perform assays of blood-brain barrier function with shear stress, with 48 independent assays simultaneously. Using the high throughput assay, we conducted drug screening assays to explore the effects of compounds for opening or closing blood-brain barrier. Our studies revealed that assays with shear stress were more predictive and were able to identify compounds known to modify the blood-brain barrier function while static assays were not. Overall, we demonstrate an optimized, high throughput assay for simulating the blood-brain barrier that incorporates shear stress and is practical for use in drug screening and other high throughput studies of toxicology.
Collapse
Affiliation(s)
- Daniel Chavarria
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Ali Abbaspour
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Natalie Celestino
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Nehali Shah
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | | | - Aaron B. Baker
- Author to whom correspondence should be addressed:. Tel.:+512-232-7114
| |
Collapse
|
9
|
Singh S, Agrawal M, Vashist R, Patel RK, Sangave SD, Alexander A. Recent advancements on in vitro blood-brain barrier model: A reliable and efficient screening approach for preclinical and clinical investigation. Expert Opin Drug Deliv 2023; 20:1839-1857. [PMID: 38100459 DOI: 10.1080/17425247.2023.2295940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION The efficiency of brain therapeutics is greatly hindered by the blood-brain barrier (BBB). BBB's protective function, selective permeability, and dynamic functionality maintain the harmony between the brain and peripheral region. Thus, the design of any novel drug carrier system requires the complete study and investigation of BBB permeability, efflux transport, and the effect of associated cellular and non-vascular unit trafficking on BBB penetrability. The in vitro BBB models offer a most promising, and reliable mode of initial investigation of BBB permeability and associated factors as strong evidence for further preclinical and clinical investigation. AREA COVERED This review work covers the structure and functions of BBB components and different types of in vitro BBB models along with factors affecting BBB model development and model selection criteria. EXPERT OPINION In vivo models assume to reciprocate the physiological environment to the maximum extent. However, the interspecies variability, NVUs trafficking, dynamic behavior of BBB, etc., lead to non-reproducible results. The in vitro models are comparatively less complex, and flexible, as per the study design, could generate substantial evidence and help identify suitable in vivo animal model selection.
Collapse
Affiliation(s)
- Snigdha Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Mukta Agrawal
- School of Pharmacy and Technology Management, Narsee Monjee Institute of Management Studies, Mahbubnagar, India
| | - Rajat Vashist
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Rohit K Patel
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | | | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| |
Collapse
|
10
|
Mármol I, Abizanda-Campo S, Ayuso JM, Ochoa I, Oliván S. Towards Novel Biomimetic In Vitro Models of the Blood-Brain Barrier for Drug Permeability Evaluation. Bioengineering (Basel) 2023; 10:bioengineering10050572. [PMID: 37237642 DOI: 10.3390/bioengineering10050572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Current available animal and in vitro cell-based models for studying brain-related pathologies and drug evaluation face several limitations since they are unable to reproduce the unique architecture and physiology of the human blood-brain barrier. Because of that, promising preclinical drug candidates often fail in clinical trials due to their inability to penetrate the blood-brain barrier (BBB). Therefore, novel models that allow us to successfully predict drug permeability through the BBB would accelerate the implementation of much-needed therapies for glioblastoma, Alzheimer's disease, and further disorders. In line with this, organ-on-chip models of the BBB are an interesting alternative to traditional models. These microfluidic models provide the necessary support to recreate the architecture of the BBB and mimic the fluidic conditions of the cerebral microvasculature. Herein, the most recent advances in organ-on-chip models for the BBB are reviewed, focusing on their potential to provide robust and reliable data regarding drug candidate ability to reach the brain parenchyma. We point out recent achievements and challenges to overcome in order to advance in more biomimetic in vitro experimental models based on OOO technology. The minimum requirements that should be met to be considered biomimetic (cellular types, fluid flow, and tissular architecture), and consequently, a solid alternative to in vitro traditional models or animals.
Collapse
Affiliation(s)
- Inés Mármol
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Sara Abizanda-Campo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Department of Dermatology, Department of Biomedical Engineering, and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jose M Ayuso
- Department of Dermatology, Department of Biomedical Engineering, and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
11
|
Foreman KL, Shusta EV, Palecek SP. Defined Differentiation of Human Pluripotent Stem Cells to Brain Microvascular Endothelial-Like Cells for Modeling the Blood-Brain Barrier. Methods Mol Biol 2023; 2683:113-133. [PMID: 37300771 PMCID: PMC10389759 DOI: 10.1007/978-1-0716-3287-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The blood-brain barrier (BBB) comprises brain microvascular endothelial cells (BMECs) that form a high-resistance cellular interface that separates the blood compartment from the brain parenchyma. An intact BBB is pivotal to maintaining brain homeostasis but also impedes the entry of neurotherapeutics. There are limited options for human-specific BBB permeability testing, however. Human pluripotent stem cell models offer a powerful tool for dissecting components of this barrier in vitro, including understanding mechanisms of BBB function, and developing strategies to improve the permeability of molecular and cellular therapeutics targeting the brain. Here, we provide a detailed, step-by-step protocol for differentiation of human pluripotent stem cells (hPSCs) to cells exhibiting key characteristics of BMECs, including paracellular and transcellular transport resistance and transporter function that enable modeling the human BBB.
Collapse
Affiliation(s)
- Koji L Foreman
- Department of Chemical and Biological Engineering, Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA.
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
12
|
Hasannejad-Asl B, Pooresmaeil F, Choupani E, Dabiri M, Behmardi A, Fadaie M, Fathi M, Moosavi SA, Takamoli S, Hemati E, Naei VY, Kazemi-Lomedasht F. Nanoparticles as Powerful Tools for Crossing the Blood-brain Barrier. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:18-26. [PMID: 35196974 DOI: 10.2174/1871527321666220222092655] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/06/2022] [Accepted: 01/16/2022] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier (BBB) is considered an important protective barrier in the central nervous system (CNS). The barrier is mainly formed by endothelial cells (ECs) interconnected by various junctions such as tight junctions (TJs), gap junctions, and adherent junctions. They collectively constitute an intensive barrier to the transit of different substances into the brain, selectively permitting small molecules to pass through by passive movement but holding off large ones such as peptides and proteins to cross the brain. Hence some molecules selectively transfer across the BBB by active routes via transcytosis. The BBB also forms a barrier against neurotoxins as well as pathogenic agents. Although various CNS disorders like Alzheimer's disease (AD) and Parkinson's disease (PD) could hamper the integrity of the border. Nevertheless, the BBB acts as a barrier for CNS disorders treatment because it prevents the drugs from reaching their target in the CNS. In recent years, different strategies, including osmotic disruption of BBB or chemical modification of drugs, have been used to transfer the chemotherapeutic agents into brain substances. Nowadays, nanoparticles (NPs) have been used as an effective and non-invasive tool for drug delivery and diagnosis of CNS disorders. In this review, we discuss the structural characteristic of BBB, safe passageways to cross the BBB, and the relation of barrier lesions with different CNS disorders. In the end, we explore the progress in drug delivery, diagnosis, imaging, and treatment of CNS disorders using nanoparticles.
Collapse
Affiliation(s)
- Behnam Hasannejad-Asl
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farkhondeh Pooresmaeil
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Edris Choupani
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mehran Dabiri
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Abtin Behmardi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mahmood Fadaie
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Majid Fathi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Seyed Akbar Moosavi
- Department of Medical Laboratory Sciences, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Neuroscience Research Center (NRC) The Iran University of Medical Science, Allied Health Department, Tehran, Iran
- Tehran Women Hospital, AST Genetic Lab, Tehran, Iran
| | - Shahla Takamoli
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Ehsan Hemati
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Yaghoubi Naei
- Immunology Research Center, Mashhad University of Medical Science, Mashhad, Iran
| | - Fatemeh Kazemi-Lomedasht
- Department of Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
13
|
Human mini-blood-brain barrier models for biomedical neuroscience research: a review. Biomater Res 2022; 26:82. [PMID: 36527159 PMCID: PMC9756735 DOI: 10.1186/s40824-022-00332-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
The human blood-brain barrier (BBB) is a unique multicellular structure that is in critical demand for fundamental neuroscience studies and therapeutic evaluation. Despite substantial achievements in creating in vitro human BBB platforms, challenges in generating specifics of physiopathological relevance are viewed as impediments to the establishment of in vitro models. In this review, we provide insight into the development and deployment of in vitro BBB models that allow investigation of the physiology and pathology of neurological therapeutic avenues. First, we highlight the critical components, including cell sources, biomaterial glue collections, and engineering techniques to reconstruct a miniaturized human BBB. Second, we describe recent breakthroughs in human mini-BBBs for investigating biological mechanisms in neurology. Finally, we discuss the application of human mini-BBBs to medical approaches. This review provides strategies for understanding neurological diseases, a validation model for drug discovery, and a potential approach for generating personalized medicine.
Collapse
|
14
|
Kawakita S, Mandal K, Mou L, Mecwan MM, Zhu Y, Li S, Sharma S, Hernandez AL, Nguyen HT, Maity S, de Barros NR, Nakayama A, Bandaru P, Ahadian S, Kim HJ, Herculano RD, Holler E, Jucaud V, Dokmeci MR, Khademhosseini A. Organ-On-A-Chip Models of the Blood-Brain Barrier: Recent Advances and Future Prospects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201401. [PMID: 35978444 PMCID: PMC9529899 DOI: 10.1002/smll.202201401] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/22/2022] [Indexed: 05/09/2023]
Abstract
The human brain and central nervous system (CNS) present unique challenges in drug development for neurological diseases. One major obstacle is the blood-brain barrier (BBB), which hampers the effective delivery of therapeutic molecules into the brain while protecting it from blood-born neurotoxic substances and maintaining CNS homeostasis. For BBB research, traditional in vitro models rely upon Petri dishes or Transwell systems. However, these static models lack essential microenvironmental factors such as shear stress and proper cell-cell interactions. To this end, organ-on-a-chip (OoC) technology has emerged as a new in vitro modeling approach to better recapitulate the highly dynamic in vivo human brain microenvironment so-called the neural vascular unit (NVU). Such BBB-on-a-chip models have made substantial progress over the last decade, and concurrently there has been increasing interest in modeling various neurological diseases such as Alzheimer's disease and Parkinson's disease using OoC technology. In addition, with recent advances in other scientific technologies, several new opportunities to improve the BBB-on-a-chip platform via multidisciplinary approaches are available. In this review, an overview of the NVU and OoC technology is provided, recent progress and applications of BBB-on-a-chip for personalized medicine and drug discovery are discussed, and current challenges and future directions are delineated.
Collapse
Affiliation(s)
- Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Lei Mou
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong, 510150, P. R. China
| | | | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Praveen Bandaru
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Rondinelli Donizetti Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
- Department of Bioprocess and Biotechnology Engineering, School of Pharmaceutical Sciences, São Paulo State University (Unesp), Araraquara, SP, 14801-902, Brazil
| | - Eggehard Holler
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90064, USA
| |
Collapse
|
15
|
Schreiner TG, Creangă-Murariu I, Tamba BI, Lucanu N, Popescu BO. In Vitro Modeling of the Blood–Brain Barrier for the Study of Physiological Conditions and Alzheimer’s Disease. Biomolecules 2022; 12:biom12081136. [PMID: 36009030 PMCID: PMC9405874 DOI: 10.3390/biom12081136] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
The blood–brain barrier (BBB) is an essential structure for the maintenance of brain homeostasis. Alterations to the BBB are linked with a myriad of pathological conditions and play a significant role in the onset and evolution of neurodegenerative diseases, including Alzheimer’s disease. Thus, a deeper understanding of the BBB’s structure and function is mandatory for a better knowledge of neurodegenerative disorders and the development of effective therapies. Because studying the BBB in vivo imposes overwhelming difficulties, the in vitro approach remains the main possible way of research. With many in vitro BBB models having been developed over the last years, the main aim of this review is to systematically present the most relevant designs used in neurological research. In the first part of the article, the physiological and structural–functional parameters of the human BBB are detailed. Subsequently, available BBB models are presented in a comparative approach, highlighting their advantages and limitations. Finally, the new perspectives related to the study of Alzheimer’s disease with the help of novel devices that mimic the in vivo human BBB milieu gives the paper significant originality.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Neurology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
- Correspondence:
| | - Ioana Creangă-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str., No. 16, 700155 Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str., No. 16, 700155 Iasi, Romania
| | - Nicolae Lucanu
- Department of Applied Electronics and Intelligent Systems, Faculty of Electronics, Telecommunications and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
16
|
Zhao N, Guo Z, Kulkarni S, Norman D, Zhang S, Chung TD, Nerenberg RF, Linville R, Searson P. Engineering the human blood-brain barrier at the capillary scale using a double-templating technique. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2110289. [PMID: 36312050 PMCID: PMC9610437 DOI: 10.1002/adfm.202110289] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Indexed: 05/15/2023]
Abstract
In vitro blood-brain barrier (BBB) models have played an important role in studying processes such as immune cell trafficking and drug delivery, as well as contributing to the understanding of mechanisms of disease progression. Many biological and pathological processes in the cerebrovasculature occur in capillaries and hence the lack of robust hierarchical models at the capillary scale is a major roadblock in BBB research. Here we report on a double-templating technique for engineering hierarchical BBB models with physiological barrier function at the capillary scale. We first demonstrate the formation of hierarchical vascular networks using human umbilical vein endothelial cells. We then characterize barrier function in a BBB model using brain microvascular endothelial-like cells (iBMECs) differentiated from induced pluripotent stem cells (iPSCs). Finally, we characterize immune cell adhesion and transmigration in response to perfusion with the inflammatory cytokine tumor necrosis factor-alpha, and show that we can recapitulate capillary-scale effects, such as leukocyte plugging, observed in mouse models. Our double-templated hierarchical model enables the study of a wide range of biological and pathological processes related to the human BBB.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Sarah Kulkarni
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Danielle Norman
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Sophia Zhang
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Tracy D. Chung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Renée F. Nerenberg
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Raleigh Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
17
|
Vetter VC, Wagner E. Targeting nucleic acid-based therapeutics to tumors: Challenges and strategies for polyplexes. J Control Release 2022; 346:110-135. [PMID: 35436520 DOI: 10.1016/j.jconrel.2022.04.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 12/18/2022]
Abstract
The current medical reality of cancer gene therapy is reflected by more than ten approved products on the global market, including oncolytic and other viral vectors and CAR T-cells as ex vivo gene-modified cell therapeutics. The development of synthetic antitumoral nucleic acid therapeutics has been proceeding at a lower but steady pace, fueled by a plethora of alternative nucleic acid platforms (from various antisense oligonucleotides, siRNA, microRNA, lncRNA, sgRNA, to larger mRNA and DNA) and several classes of physical and chemical delivery technologies. This review summarizes the challenges and strategies for tumor-targeted nucleic acid delivery. Focusing primarily on polyplexes (polycation complexes) as nanocarriers, delivery options across multiple barriers into tumor cells are illustrated.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Ludwig-Maximilians-Universität, Munich 81377, Germany; Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Munich 81377, Germany.
| |
Collapse
|
18
|
Fengler S, Kurkowsky B, Kaushalya SK, Roth W, Fava E, Denner P. Human iPSC-derived brain endothelial microvessels in a multi-well format enable permeability screens of anti-inflammatory drugs. Biomaterials 2022; 286:121525. [DOI: 10.1016/j.biomaterials.2022.121525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 03/31/2022] [Accepted: 04/11/2022] [Indexed: 12/27/2022]
|
19
|
Beth Payne L, Tewari BP, Dunkenberger L, Bond S, Savelli A, Darden J, Zhao H, Willi C, Kanodia R, Gude R, Powell MD, Oestreich KJ, Sontheimer H, Dal-Pra S, Chappell JC. Pericyte Progenitor Coupling to the Emerging Endothelium During Vasculogenesis via Connexin 43. Arterioscler Thromb Vasc Biol 2022; 42:e96-e114. [PMID: 35139658 PMCID: PMC8957572 DOI: 10.1161/atvbaha.121.317324] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/24/2022] [Indexed: 01/23/2023]
Abstract
BACKGROUND Vascular pericytes stabilize blood vessels and contribute to their maturation, while playing other key roles in microvascular function. Nevertheless, relatively little is known about involvement of their precursors in the earliest stages of vascular development, specifically during vasculogenesis. METHODS We combined high-power, time-lapse imaging with transcriptional profiling of emerging pericytes and endothelial cells in reporter mouse and cell lines. We also analyzed conditional transgenic animals deficient in Cx43/Gja1 (connexin 43/gap junction alpha-1) expression within Ng2+ cells. RESULTS A subset of Ng2-DsRed+ cells, likely pericyte/mural cell precursors, arose alongside endothelial cell differentiation and organization and physically engaged vasculogenic endothelium in vivo and in vitro. We found no overlap between this population of differentiating pericyte/mural progenitors and other lineages including hemangiogenic and neuronal/glial cell types. We also observed cell-cell coupling and identified Cx43-based gap junctions contributing to pericyte-endothelial cell precursor communication during vascular assembly. Genetic loss of Cx43/Gja1 in Ng2+ pericyte progenitors compromised embryonic blood vessel formation in a subset of animals, while surviving mutants displayed little-to-no vessel abnormalities, suggesting a resilience to Cx43/Gja1 loss in Ng2+ cells or potential compensation by additional connexin isoforms. CONCLUSIONS Together, our data suggest that a distinct pericyte lineage emerges alongside vasculogenesis and directly communicates with the nascent endothelium via Cx43 during early vessel formation. Cx43/Gja1 loss in pericyte/mural cell progenitors can induce embryonic vessel dysmorphogenesis, but alternate connexin isoforms may be able to compensate. These data provide insight that may reshape the current framework of vascular development and may also inform tissue revascularization/vascularization strategies.
Collapse
Affiliation(s)
- Laura Beth Payne
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Bhanu P. Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903, USA
| | - Logan Dunkenberger
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Samantha Bond
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Alyssa Savelli
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Jordan Darden
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA 24061, USA
| | - Huaning Zhao
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Caroline Willi
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Ronak Kanodia
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Rosalie Gude
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Michael D. Powell
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kenneth J. Oestreich
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903, USA
| | - Sophie Dal-Pra
- Division of Cardiovascular Medicine and Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - John C. Chappell
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
20
|
Shah B, Dong X. Current Status of In vitro Models of the Blood-Brain Barrier. Curr Drug Deliv 2022; 19:1034-1046. [PMID: 35240972 DOI: 10.2174/1567201819666220303102614] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/04/2022] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
Disorders of the brain are the most debilitating situation affected globally with increased mortality rates every year, while brain physiology and cumbersome drug development processes exacerbate this. Although blood-brain barrier (BBB) and its components are important for brain protection, their complexity creates major obstacles for brain drug delivery and the BBB is the primary cause of treatment failure leading to disease progression. Therefore, developing an ideal platform which can predict the behavior of a drug delivery system in the brain at the early development phase is extremely crucial. In this direction, since the last two decades, numerous in vitro BBB models have been developed and investigated by researchers to understand the barrier properties and how closely the in vitro models mimic with in vivo BBB. In-vitro BBB models are mainly culture of endothelial cells or their coculture with other perivascular cells either in two or three-dimensional platforms. In this article, we have briefly summarized the fundamentals of BBB and outlined different types of in vitro BBB models with their pros and cons. Based on the available reports, no model seems to be robust that can truly mimic the entire properties of the in vivo BBB microvasculature. However, human stem cells, coculture and three-dimensional models were found to mimic the complexity of the barrier integrity not completely but more precisely than other in vitro models. More studies aiming towards combining them together would be needed to develop an ideal in vitro model that can overcome the existing limitations and unravel the mysterious BBB.
Collapse
Affiliation(s)
- Brijesh Shah
- Department of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Xiaowei Dong
- Department of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, USA
| |
Collapse
|
21
|
Yang PH, Zheng FY, Li QS, Tian T, Zhang GY, Wu L, Mao HJ. An easy-repeat method to build a blood-brain barrier model on a chip with independent TEER detection module. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2022. [DOI: 10.1016/j.cjac.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Newman Frisch A, Debbi L, Shuhmaher M, Guo S, Levenberg S. Advances in vascularization and innervation of constructs for neural tissue engineering. Curr Opin Biotechnol 2022; 73:188-197. [PMID: 34481245 DOI: 10.1016/j.copbio.2021.08.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 02/05/2023]
Abstract
A growing number of technologies are being developed to promote vascularization and innervation in engineered tissues. Organ-on-a-chip, organoid and 3D printing technologies, as well as pre-vascularized and oriented scaffolds, have been employed for vascularization and innervation of engineered tissues both in vivo and in vitro. Both vascularization and innervation are critical for neural tissue engineering, as these complex tissues require provision of both blood and nerves. As such, this review will have particular focus on neural tissue engineering. We examine state-of-the-art approaches for tissue vascularization and innervation and identify promising methods for developing vascularized and innervated engineered neural constructs.
Collapse
Affiliation(s)
- Abigail Newman Frisch
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Lior Debbi
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Margarita Shuhmaher
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Shaowei Guo
- The First Affiliated Hospital, Shantou University Medical College, Shantou 515000, China
| | - Shulamit Levenberg
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
23
|
Martins C, Sarmento B. Cell Interplay Model to Assess the Impact of Glioma Cells on Blood-Brain Barrier Permeability. Methods Mol Biol 2022; 2492:267-276. [PMID: 35733050 DOI: 10.1007/978-1-0716-2289-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) is the most selective protecting layer of the central nervous system (CNS) with unique neurovascular features. The BBB is known to undergo a process of molecular alterations during disease state, such as in the case of glioma. This results in a non-uniform permeability along the BBB layer, which retains intact regions but develops focal sites of higher leakiness, especially in the surrounds of the tumor core. Although essential to guarantee brain homeostasis, the BBB has been the Achilles heel of drug delivery to the brain since the early times of the first classification as "barrier," more than a century ago. Due to the presence of the BBB, the transport of drug molecules from the bloodstream to the brain parenchyma is highly restricted, and, therefore, clinically relevant therapeutic concentrations cannot be achieved. Research efforts have focused on the development of novel tools to ameliorate drug permeability across the BBB, including drug formulation into non-invasive delivery systems with brain targeting properties and techniques that allow a temporary disruption of the BBB. To strengthen the advancement of potential drug candidates, in vitro models that recapitulate the main in vivo features of BBB are required to perform a preliminary screening of permeability, both in health and disease conditions. Herein, a protocol to assemble a BBB in vitro model to screen drug permeability in a glioma disease state is detailed. The model consists of a BBB and glioma cell co-culture and aims at exploiting the effect of the interplay between the cell constituents on the permeability of drug molecules. Although simple and straightforward, the herein in vitro model presents a high reproducibility, cost-effectiveness, and a favorable time-benefit balance.
Collapse
Affiliation(s)
- Cláudia Martins
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Bruno Sarmento
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.
- Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde (CESPU), Gandra, Portugal.
| |
Collapse
|
24
|
Potjewyd G, Kellett K, Hooper N. 3D hydrogel models of the neurovascular unit to investigate blood-brain barrier dysfunction. Neuronal Signal 2021; 5:NS20210027. [PMID: 34804595 PMCID: PMC8579151 DOI: 10.1042/ns20210027] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 12/16/2022] Open
Abstract
The neurovascular unit (NVU), consisting of neurons, glial cells, vascular cells (endothelial cells, pericytes and vascular smooth muscle cells (VSMCs)) together with the surrounding extracellular matrix (ECM), is an important interface between the peripheral blood and the brain parenchyma. Disruption of the NVU impacts on blood-brain barrier (BBB) regulation and underlies the development and pathology of multiple neurological disorders, including stroke and Alzheimer's disease (AD). The ability to differentiate induced pluripotent stem cells (iPSCs) into the different cell types of the NVU and incorporate them into physical models provides a reverse engineering approach to generate human NVU models to study BBB function. To recapitulate the in vivo situation such NVU models must also incorporate the ECM to provide a 3D environment with appropriate mechanical and biochemical cues for the cells of the NVU. In this review, we provide an overview of the cells of the NVU and the surrounding ECM, before discussing the characteristics (stiffness, functionality and porosity) required of hydrogels to mimic the ECM when incorporated into in vitro NVU models. We summarise the approaches available to measure BBB functionality and present the techniques in use to develop robust and translatable models of the NVU, including transwell models, hydrogel models, 3D-bioprinting, microfluidic models and organoids. The incorporation of iPSCs either without or with disease-specific genetic mutations into these NVU models provides a platform in which to study normal and disease mechanisms, test BBB permeability to drugs, screen for new therapeutic targets and drugs or to design cell-based therapies.
Collapse
Affiliation(s)
- Geoffrey Potjewyd
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Katherine A.B. Kellett
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Nigel M. Hooper
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, U.K
| |
Collapse
|
25
|
Yan L, Moriarty RA, Stroka KM. Recent progress and new challenges in modeling of human pluripotent stem cell-derived blood-brain barrier. Theranostics 2021; 11:10148-10170. [PMID: 34815809 PMCID: PMC8581424 DOI: 10.7150/thno.63195] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
The blood-brain barrier (BBB) is a semipermeable unit that serves to vascularize the central nervous system (CNS) while tightly regulating the movement of molecules, ions, and cells between the blood and the brain. The BBB precisely controls brain homeostasis and protects the neural tissue from toxins and pathogens. The BBB is coordinated by a tight monolayer of brain microvascular endothelial cells, which is subsequently supported by mural cells, astrocytes, and surrounding neuronal cells that regulate the barrier function with a series of specialized properties. Dysfunction of barrier properties is an important pathological feature in the progression of various neurological diseases. In vitro BBB models recapitulating the physiological and diseased states are important tools to understand the pathological mechanism and to serve as a platform to screen potential drugs. Recent advances in this field have stemmed from the use of pluripotent stem cells (PSCs). Various cell types of the BBB such as brain microvascular endothelial cells (BMECs), pericytes, and astrocytes have been derived from PSCs and synergistically incorporated to model the complex BBB structure in vitro. In this review, we summarize the most recent protocols and techniques for the differentiation of major cell types of the BBB. We also discuss the progress of BBB modeling by using PSC-derived cells and perspectives on how to reproduce more natural BBBs in vitro.
Collapse
Affiliation(s)
- Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Rebecca A. Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Biophysics Program, University of Maryland, College Park, MD 20742, USA
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
26
|
Augustine R, Aqel AH, Kalva SN, Joshy KS, Nayeem A, Hasan A. Bioengineered microfluidic blood-brain barrier models in oncology research. Transl Oncol 2021; 14:101087. [PMID: 33865030 PMCID: PMC8066424 DOI: 10.1016/j.tranon.2021.101087] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/25/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Metastasis is the major reason for most brain tumors with up to a 50% chance of occurrence in patients with other types of malignancies. Brain metastasis occurs if cancer cells succeed to cross the 'blood-brain barrier' (BBB). Moreover, changes in the structure and function of BBB can lead to the onset and progression of diseases including neurological disorders and brain-metastases. Generating BBB models with structural and functional features of intact BBB is highly important to better understand the molecular mechanism of such ailments and finding novel therapeutic agents targeting them. Hence, researchers are developing novel in vitro BBB platforms that can recapitulate the structural and functional characteristics of BBB. Brain endothelial cells-based in vitro BBB models have thus been developed to investigate the mechanism of brain metastasis through BBB and facilitate the testing of brain targeted anticancer drugs. Bioengineered constructs integrated with microfluidic platforms are vital tools for recapitulating the features of BBB in vitro closely as possible. In this review, we outline the fundamentals of BBB biology, recent developments in the microfluidic BBB platforms, and provide a concise discussion of diverse types of bioengineered BBB models with an emphasis on the application of them in brain metastasis and cancer research in general. We also provide insights into the challenges and prospects of the current bioengineered microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| | - Ahmad H Aqel
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Sumama Nuthana Kalva
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - K S Joshy
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur 680020, Kerala, India
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| |
Collapse
|
27
|
Gupta R, Sharma D. Therapeutic response differences between 2D and 3D tumor models of magnetic hyperthermia. NANOSCALE ADVANCES 2021; 3:3663-3680. [PMID: 36133021 PMCID: PMC9418625 DOI: 10.1039/d1na00224d] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/05/2021] [Indexed: 05/02/2023]
Abstract
Magnetic hyperthermia-based cancer therapy (MHCT) has surfaced as one of the promising techniques for inaccessible solid tumors. It involves generation of localized heat in the tumor tissues on application of an alternating magnetic field in the presence of magnetic nanoparticles (MNPs). Unfortunately, lack of precise temperature and adequate MNP distribution at the tumor site under in vivo conditions has limited its application in the biomedical field. Evaluation of in vitro tumor models is an alternative for in vivo models. However, generally used in vitro two-dimensional (2D) models cannot mimic all the characteristics of a patient's tumor and hence, fail to establish or address the experimental variables and concerns. Considering that three-dimensional (3D) models have emerged as the best possible state to replicate the in vivo conditions successfully in the laboratory for most cell types, it is possible to conduct MHCT studies with higher clinical relevance for the analysis of the selection of magnetic parameters, MNP distribution, heat dissipation, action and acquired thermotolerance in cancer cells. In this review, various forms of 3D cultures have been considered and the successful implication of MHCT on them has been summarized, which includes tumor spheroids, and cultures grown in scaffolds, cell culture inserts and microfluidic devices. This review aims to summarize the contrast between 2D and 3D in vitro tumor models for pre-clinical MHCT studies. Furthermore, we have collated and discussed the usefulness, suitability, pros and cons of these tumor models. Even though numerous cell culture models have been established, further investigations on the new pre-clinical models and selection of best fit model for successful MHCT applications are still necessary to confer a better understanding for researchers.
Collapse
Affiliation(s)
- Ruby Gupta
- Institute of Nano Science and Technology Knowledge City, Sector 81 Mohali Punjab-140306 India
| | - Deepika Sharma
- Institute of Nano Science and Technology Knowledge City, Sector 81 Mohali Punjab-140306 India
| |
Collapse
|
28
|
Wasson EM, Dubbin K, Moya ML. Go with the flow: modeling unique biological flows in engineered in vitro platforms. LAB ON A CHIP 2021; 21:2095-2120. [PMID: 34008661 DOI: 10.1039/d1lc00014d] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Interest in recapitulating in vivo phenomena in vitro using organ-on-a-chip technology has grown rapidly and with it, attention to the types of fluid flow experienced in the body has followed suit. These platforms offer distinct advantages over in vivo models with regards to human relevance, cost, and control of inputs (e.g., controlled manipulation of biomechanical cues from fluid perfusion). Given the critical role biophysical forces play in several tissues and organs, it is therefore imperative that engineered in vitro platforms capture the complex, unique flow profiles experienced in the body that are intimately tied with organ function. In this review, we outline the complex and unique flow regimes experienced by three different organ systems: blood vasculature, lymphatic vasculature, and the intestinal system. We highlight current state-of-the-art platforms that strive to replicate physiological flows within engineered tissues while introducing potential limitations in current approaches.
Collapse
Affiliation(s)
- Elisa M Wasson
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Karen Dubbin
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Monica L Moya
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| |
Collapse
|
29
|
Bisphenol A Inhibits the Transporter Function of the Blood-Brain Barrier by Directly Interacting with the ABC Transporter Breast Cancer Resistance Protein (BCRP). Int J Mol Sci 2021; 22:ijms22115534. [PMID: 34073890 PMCID: PMC8197233 DOI: 10.3390/ijms22115534] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
The breast cancer resistance protein (BCRP) is an important efflux transporter in the blood-brain barrier (BBB), protecting the brain from a wide range of substances. In this study, we investigated if BCRP function is affected by bisphenol A (BPA), a high production volume chemical used in common consumer products, as well as by bisphenol F (BPF) and bisphenol S (BPS), which are used to substitute BPA. We employed a transwell-based in vitro cell model of iPSC-derived brain microvascular endothelial cells, where BCRP function was assessed by measuring the intracellular accumulation of its substrate Hoechst 33342. Additionally, we used in silico modelling to predict if the bisphenols could directly interact with BCRP. Our results showed that BPA significantly inhibits the transport function of BCRP. Additionally, BPA was predicted to bind to the cavity that is targeted by known BCRP inhibitors. Taken together, our findings demonstrate that BPA inhibits BCRP function in vitro, probably by direct interaction with the transporter. This effect might contribute to BPA’s known impact on neurodevelopment.
Collapse
|
30
|
Nozohouri S, Noorani B, Al-Ahmad A, Abbruscato TJ. Estimating Brain Permeability Using In Vitro Blood-Brain Barrier Models. Methods Mol Biol 2021; 2367:47-72. [PMID: 32789777 DOI: 10.1007/7651_2020_311] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The blood-brain barrier (BBB) is a vital biological interface that regulates transfer of different molecules between blood and brain and, therefore, maintains the homeostatic environment of the CNS. In order to perform high-throughput screening of therapeutics in drug discovery, specific properties of the BBB are investigated within in vitro BBB platforms. In this chapter, we detail the process and steps for the iPSC to BMEC and astrocyte differentiation as well as TEER and permeability measurement in Transwell platform of in vitro BBB model. Also, advanced microfluidic iPSCs-derived BMECs on chip and permeability measurement within this model have been elucidated.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
- Center for Blood-Brain Barrier Research, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
31
|
Wang H, Xu X, Yin Y, Yu S, Ren H, Xue Q, Xu X. Catalpol protects vascular structure and promotes angiogenesis in cerebral ischemic rats by targeting HIF-1α/VEGF. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153300. [PMID: 32866905 DOI: 10.1016/j.phymed.2020.153300] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/01/2020] [Accepted: 08/10/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The initial factor in the occurrence, development, and prognosis of cerebral ischemia is vascular dysfunction in the brain, and vascular remodeling of the brain is the key therapeutic target and strategy for ischemic tissue repair. Catalpol is the main active component of the radix of Rehmannia glutinosa Libosch, and it exhibits potential pleiotropic protective effects in many brain-related diseases, including stroke. PURPOSE The present study was designed to investigate whether catalpol protects vascular structure and promotes angiogenesis in cerebral ischemic rats and to identify its possible mechanisms in vivo and in vitro. STUDY DESIGN Cerebral ischemic rats and oxygen-glucose deprivation-exposed brain microvascular endothelial cells were used to study the therapeutic potential of catalpol in vivo and in vitro. METHODS First, neurological deficits, histopathological morphology, infarct volume, vascular morphology, vessel density, and angiogenesis in focal cerebral ischemic rats were observed to test the potential treatment effects of catalpol. Then, oxygen-glucose deprivation-exposed brain microvascular endothelial cells were used to mimic the pathological changes in vessels during ischemia to study the effects and possible mechanisms of catalpol in protecting vascular structure and promoting angiogenesis. RESULTS The in vivo results showed that catalpol reduced neurological deficit scores and infarct volume, protected vascular structure, and promoted angiogenesis in cerebral ischemic rats. The in vitro results showed that catalpol improved oxygen-glucose deprivation-induced damage and promoted proliferation, migration, and in vitro tube formation of brain microvascular endothelial cells. The HIF-1α (hypoxia-inducible factor 1α)/VEGF (vascular endothelial growth factor) pathway was activated by catalpol both in the brains of cerebral ischemic rats and in primary brain microvascular endothelial cells, and the activating effects of catalpol were inhibited by SU1498. CONCLUSION The results of both the in vivo and in vitro studies proved that catalpol protects vascular structure and promotes angiogenesis in focal cerebral ischemic rats and that the mechanism is dependent on HIF-1α/VEGF.
Collapse
Affiliation(s)
- Hongjin Wang
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Xiaogang Xu
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Yue Yin
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Shiqi Yu
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Huijing Ren
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Qiang Xue
- Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences & Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China; Southwest University Hospital, Chongqing 400715, China.
| |
Collapse
|
32
|
Meenambal R, Srinivas Bharath MM. Nanocarriers for effective nutraceutical delivery to the brain. Neurochem Int 2020; 140:104851. [PMID: 32976906 DOI: 10.1016/j.neuint.2020.104851] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
Neurodegenerative disorders are common among aging populations around the globe. Most are characterized by loss of neurons, protein aggregates, oxidative stress, mitochondrial damage, neuroinflammation among others. Although symptomatic treatment using conventional pharmacotherapy has been widely employed, their therapeutic success is limited due to varied reasons. In the need to identify an alternative approach, researchers successfully demonstrated the therapeutic utility of plant-derived nutraceuticals in cell and animal models of neurodegenerative conditions. However, most nutraceuticals failed during clinical trials in humans owing to their poor bioavailability in vivo and limited permeability across the blood brain barrier (BBB). The current emphasis is therefore on the improved delivery of nutraceuticals to the brain. In this regard, development of nanoparticle conjugated nutraceuticals to enhance bioavailability and therapeutic efficacy in the brain has gained attention. Here, we review the research advances in nanoparticles conjugated nutraceuticals applied in neurodegenerative disorders and discuss their advantages and limitations, clinical trials and toxicity concerns.
Collapse
Affiliation(s)
- Rugmani Meenambal
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India.
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India; Neurotoxicology Laboratory, Neurobiology Research Centre, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India.
| |
Collapse
|
33
|
Ille AM, Kishel E, Bodea R, Ille A, Lamont H, Amico-Ruvio S. Protein LY6E as a candidate for mediating transport of adeno-associated virus across the human blood-brain barrier. J Neurovirol 2020; 26:769-778. [PMID: 32839948 DOI: 10.1007/s13365-020-00890-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 07/14/2020] [Accepted: 08/04/2020] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) is a major obstacle for the treatment of central nervous system (CNS) disorders. Significant progress has been made in developing adeno-associated virus (AAV) variants with increased ability to cross the BBB in mice. However, these variants are not efficacious in non-human primates. Herein, we employed various bioinformatic techniques to identify lymphocyte antigen-6E (LY6E) as a candidate for mediating transport of AAV across the human BBB based on the previously determined mechanism of transport in mice. Our results provide insight into future discovery and optimization of AAV variants for CNS gene delivery in humans.
Collapse
Affiliation(s)
- Alexander M Ille
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ, 07103, USA.,STEM Biomedical, Kitchener, ON, N2M 3B9, Canada
| | - Eric Kishel
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Raoul Bodea
- STEM Biomedical, Kitchener, ON, N2M 3B9, Canada
| | - Anetta Ille
- STEM Biomedical, Kitchener, ON, N2M 3B9, Canada
| | - Hannah Lamont
- Graduate School of Biomedical Sciences, Rutgers University, Newark, NJ, 07103, USA
| | | |
Collapse
|
34
|
Hynes WF, Pepona M, Robertson C, Alvarado J, Dubbin K, Triplett M, Adorno JJ, Randles A, Moya ML. Examining metastatic behavior within 3D bioprinted vasculature for the validation of a 3D computational flow model. SCIENCE ADVANCES 2020; 6:eabb3308. [PMID: 32923637 PMCID: PMC7449690 DOI: 10.1126/sciadv.abb3308] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/14/2020] [Indexed: 05/02/2023]
Abstract
Understanding the dynamics of circulating tumor cell (CTC) behavior within the vasculature has remained an elusive goal in cancer biology. To elucidate the contribution of hydrodynamics in determining sites of CTC vascular colonization, the physical forces affecting these cells must be evaluated in a highly controlled manner. To this end, we have bioprinted endothelialized vascular beds and perfused these constructs with metastatic mammary gland cells under physiological flow rates. By pairing these in vitro devices with an advanced computational flow model, we found that the bioprinted analog was readily capable of evaluating the accuracy and integrated complexity of a computational flow model, while also highlighting the discrete contribution of hydrodynamics in vascular colonization. This intersection of these two technologies, bioprinting and computational simulation, is a key demonstration in the establishment of an experimentation pipeline for the understanding of complex biophysical events.
Collapse
Affiliation(s)
- W. F. Hynes
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - M. Pepona
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - C. Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - J. Alvarado
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - K. Dubbin
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - M. Triplett
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| | - J. J. Adorno
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - A. Randles
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - M. L. Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA 94550, USA
| |
Collapse
|
35
|
Pradhan S, Banda OA, Farino CJ, Sperduto JL, Keller KA, Taitano R, Slater JH. Biofabrication Strategies and Engineered In Vitro Systems for Vascular Mechanobiology. Adv Healthc Mater 2020; 9:e1901255. [PMID: 32100473 PMCID: PMC8579513 DOI: 10.1002/adhm.201901255] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/24/2020] [Indexed: 12/17/2022]
Abstract
The vascular system is integral for maintaining organ-specific functions and homeostasis. Dysregulation in vascular architecture and function can lead to various chronic or acute disorders. Investigation of the role of the vascular system in health and disease has been accelerated through the development of tissue-engineered constructs and microphysiological on-chip platforms. These in vitro systems permit studies of biochemical regulation of vascular networks and parenchymal tissue and provide mechanistic insights into the biophysical and hemodynamic forces acting in organ-specific niches. Detailed understanding of these forces and the mechanotransductory pathways involved is necessary to develop preventative and therapeutic strategies targeting the vascular system. This review describes vascular structure and function, the role of hemodynamic forces in maintaining vascular homeostasis, and measurement approaches for cell and tissue level mechanical properties influencing vascular phenomena. State-of-the-art techniques for fabricating in vitro microvascular systems, with varying degrees of biological and engineering complexity, are summarized. Finally, the role of vascular mechanobiology in organ-specific niches and pathophysiological states, and efforts to recapitulate these events using in vitro microphysiological systems, are explored. It is hoped that this review will help readers appreciate the important, but understudied, role of vascular-parenchymal mechanotransduction in health and disease toward developing mechanotherapeutics for treatment strategies.
Collapse
Affiliation(s)
- Shantanu Pradhan
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India
| | - Omar A. Banda
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Cindy J. Farino
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John L. Sperduto
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Keely A. Keller
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - Ryan Taitano
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
| | - John H. Slater
- Department of Biomedical Engineering, University of Delaware, 150 Academy Street, 161 Colburn Lab, Newark, DE, 19716, USA
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE 19716, USA
- Delaware Biotechnology Institute, 15 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
36
|
A Reconfigurable In Vitro Model for Studying the Blood-Brain Barrier. Ann Biomed Eng 2019; 48:780-793. [PMID: 31741228 DOI: 10.1007/s10439-019-02405-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022]
Abstract
Much of what is currently known about the role of the blood-brain barrier (BBB) in regulating the passage of chemicals from the blood stream to the central nervous system (CNS) comes from animal in vivo models (requiring extrapolation to human relevance) and 2D static in vitro systems, which fail to capture the rich cell-cell and cell-matrix interactions of the dynamic 3D in vivo tissue microenvironment. In this work we have developed a BBB platform that allows for a high degree of customization in cellular composition, cellular orientation, and physiologically-relevant fluid dynamics. The system characterized and presented in this study reproduces key characteristics of a BBB model (e.g. tight junctions, efflux pumps) allowing for the formation of a selective and functional barrier. We demonstrate that our in vitro BBB is responsive to both biochemical and mechanical cues. This model further allows for culture of a CNS-like space around the BBB. The design of this platform is a valuable tool for studying BBB function as well as for screening of novel therapeutics.
Collapse
|
37
|
Chen C, Ding Q, Shen B, Yu T, Wang H, Xu Y, Guo H, Hu K, Xie L, Wang G, Liang Y. Insights into the Authentic Active Ingredients and Action Sites of Oral Exogenous Glutathione in the Treatment of Ischemic Brain Injury Based on Pharmacokinetic-Pharmacodynamic Studies. Drug Metab Dispos 2019; 48:52-62. [PMID: 31704713 DOI: 10.1124/dmd.119.089458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023] Open
Abstract
Glutathione (GSH) has been reported to be closely related to various diseases of the central nervous system, yet its authentic active ingredients and action sites remain unclear. In the present study, oral exogenous GSH significantly alleviated ischemic brain injury, but this result was inconsistent with its low bioavailability and blood-brain barrier (BBB) permeability. To ascertain the exposure of GSH-derived ingredients, including GSH, cysteine (CYS), glutamate (Glu), glycine (GLY), CYS-GLY, and γ-glutamylcysteine (γ-GC) were systematically studied both in vitro and in vivo. The outcomes demonstrated that oral GSH not only increases the GSH and CYS levels in rat striatum and cortex, but it also can decrease the rise of intracerebral Glu concentration caused by ischemia/reperfusion surgery. Then the influence of GSH on the BBB was investigated via measuring IgG leakage, intracerebral endotoxin, and tight-junction proteins. All indicators showed that GSH dosing can repair the destroyed BBB. Oral GSH greatly enhances the exposure of GSH, CYS, CYS-GLY, and γ-GC in rat duodenum, jejunum, ileum, and colon. Accumulating evidence reveals a close link between brain injury and gastrointestinal dysfunction. Our findings further suggest that oral GSH significantly improves intestinal inflammatory damage and barrier disruptions. In conclusion, oral GSH can have a direct therapeutic role in brain injury by stabilizing intracerebral levels of GSH, CYS, and Glu. It can also play an indirect therapeutic role by enhancing the intestinal exposure of GSH, CYS, CYS-GLY, and γ-GC and improving intestinal barrier disruptions. SIGNIFICANCE STATEMENT: The authentic active ingredients and action sites of exogenous glutathione (GSH) in the treatment of ischemic brain injury are unclear. We have shown that oral exogenous GSH not only stabilizes intracerebral levels of GSH, cysteine (CYS), and glutamate (Glu) to act directly on brain injury, but it can also exert an indirect therapeutic role by improving intestinal barrier disruptions. These findings have great significance for revealing the therapeutic effect of GSH on ischemic brain injury and for promoting its further development and clinical application.
Collapse
Affiliation(s)
- Chong Chen
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Qingqing Ding
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Boyu Shen
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Tengjie Yu
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - He Wang
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Yangfan Xu
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Huimin Guo
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Kangrui Hu
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Lin Xie
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| | - Yan Liang
- Key Laboratory of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University (C.C., B.S., T.Y., H.W., K.H., L.X., G.W., Y.L.), and Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University (Jiangsu People's Hospital (Q.D.), Nanjing, P.R. China
| |
Collapse
|
38
|
Roux GL, Jarray R, Guyot AC, Pavoni S, Costa N, Théodoro F, Nassor F, Pruvost A, Tournier N, Kiyan Y, Langer O, Yates F, Deslys JP, Mabondzo A. Proof-of-Concept Study of Drug Brain Permeability Between in Vivo Human Brain and an in Vitro iPSCs-Human Blood-Brain Barrier Model. Sci Rep 2019; 9:16310. [PMID: 31690750 PMCID: PMC6831611 DOI: 10.1038/s41598-019-52213-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/10/2019] [Indexed: 12/22/2022] Open
Abstract
The development of effective central nervous system (CNS) drugs has been hampered by the lack of robust strategies to mimic the blood-brain barrier (BBB) and cerebrovascular impairments in vitro. Recent technological advancements in BBB modeling using induced pluripotent stem cells (iPSCs) allowed to overcome some of these obstacles, nonetheless the pertinence for their use in drug permeation study remains to be established. This mandatory information requires a cross comparison of in vitro and in vivo pharmacokinetic data in the same species to avoid failure in late clinical drug development. Here, we measured the BBB permeabilities of 8 clinical positron emission tomography (PET) radioligands with known pharmacokinetic parameters in human brain in vivo with a newly developed in vitro iPSC-based human BBB (iPSC-hBBB) model. Our findings showed a good correlation between in vitro and in vivo drug brain permeability (R2 = 0.83; P = 0.008) which contrasted with the limited correlation between in vitro apparent permeability for a set of 18 CNS/non-CNS compounds using the in vitro iPSCs-hBBB model and drug physicochemical properties. Our data suggest that the iPSC-hBBB model can be integrated in a flow scheme of CNS drug screening and potentially used to study species differences in BBB permeation.
Collapse
Affiliation(s)
- Gwenaëlle Le Roux
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Rafika Jarray
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Anne-Cécile Guyot
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Serena Pavoni
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France
| | - Narciso Costa
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Frédéric Théodoro
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Ferid Nassor
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Alain Pruvost
- Service de Pharmacologie et d'Immunoanalyse (SPI), Plateforme Smart-MS, CEA, INRA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm, Univ. Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Yulia Kiyan
- Medizinische Hochschule Hannover, DE-30625, Hannover, Germany
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria.,Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, A-2444, Seibersdorf, Austria
| | - Frank Yates
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France.,Sup'Biotech, F-94800, Villejuif, France
| | - Jean Philippe Deslys
- Service d'Etude des Prions et des Infections Atypiques, CEA, F-92265, Fontenay-aux-Roses, France
| | - Aloïse Mabondzo
- Service de Pharmacologie et d'Immunoanalyse, CEA, Université Paris-Saclay, F-91191, Gif-sur-Yvette, France.
| |
Collapse
|
39
|
Pranda MA, Gray KM, DeCastro AJL, Dawson GM, Jung JW, Stroka KM. Tumor Cell Mechanosensing During Incorporation into the Brain Microvascular Endothelium. Cell Mol Bioeng 2019; 12:455-480. [PMID: 31719927 DOI: 10.1007/s12195-019-00591-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction Tumor metastasis to the brain occurs in approximately 20% of all cancer cases and often occurs due to tumor cells crossing the blood-brain barrier (BBB). The brain microenvironment is comprised of a soft hyaluronic acid (HA)-rich extracellular matrix with an elastic modulus of 0.1-1 kPa, whose crosslinking is often altered in disease states. Methods To explore the effects of HA crosslinking on breast tumor cell migration, we developed a biomimetic model of the human brain endothelium, consisting of brain microvascular endothelial cell (HBMEC) monolayers on HA and gelatin (HA/gelatin) films with different degrees of crosslinking, as established by varying the concentration of the crosslinker Extralink. Results and Discussion Metastatic breast tumor cell migration speed, diffusion coefficient, spreading area, and aspect ratio increased with decreasing HA crosslinking, a mechanosensing trend that correlated with tumor cell actin organization but not CD44 expression. Meanwhile, breast tumor cell incorporation into endothelial monolayers was independent of HA crosslinking density, suggesting that alterations in HA crosslinking density affect tumor cells only after they exit the vasculature. Tumor cells appeared to exploit both the paracellular and transcellular routes of trans-endothelial migration. Quantitative phenotyping of HBMEC junctions via a novel Python software revealed a VEGF-dependent decrease in punctate VE-cadherin junctions and an increase in continuous and perpendicular junctions when HBMECs were treated with tumor cell-secreted factors. Conclusions Overall, our quantitative results suggest that a combination of biochemical and physical factors promote tumor cell migration through the BBB.
Collapse
Affiliation(s)
- Marina A Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kelsey M Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Ariana Joy L DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Gregory M Dawson
- Department of Biology, University of Maryland, College Park, College Park, MD 20742 USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA.,Biophysics Program, University of Maryland, College Park, College Park, MD 20742 USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Fischell Department of Bioengineering, University of Maryland, College Park, 3110 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742 USA
| |
Collapse
|
40
|
Ambrosini YM, Borcherding D, Kanthasamy A, Kim HJ, Willette AA, Jergens A, Allenspach K, Mochel JP. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front Aging Neurosci 2019; 11:130. [PMID: 31275138 PMCID: PMC6591269 DOI: 10.3389/fnagi.2019.00130] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
Identifying appropriate animal models is critical in developing translatable in vitro and in vivo systems for therapeutic drug development and investigating disease pathophysiology. These animal models should have direct biological and translational relevance to the underlying disease they are supposed to mimic. Aging dogs not only naturally develop a cognitive decline in many aspects including learning and memory deficits, but they also exhibit human-like individual variability in the aging process. Neurodegenerative processes that can be observed in both human and canine brains include the progressive accumulation of β-amyloid (Aβ) found as diffuse plaques in the prefrontal cortex (PFC), including the gyrus proreus (i.e., medial orbital PFC), as well as the hippocampus and the cerebral vasculature. Tau pathology, a marker of neurodegeneration and dementia progression, was also found in canine hippocampal synapses. Various epidemiological data show that human patients with neurodegenerative diseases have concurrent intestinal lesions, and histopathological changes in the gastrointestinal (GI) tract occurs decades before neurodegenerative changes. Gut microbiome alterations have also been reported in many neurodegenerative diseases including Alzheimer's (AD) and Parkinson's diseases, as well as inflammatory central nervous system (CNS) diseases. Interestingly, the dog gut microbiome more closely resembles human gut microbiome in composition and functional overlap compared to rodent models. This article reviews the physiology of the gut-brain axis (GBA) and its involvement with neurodegenerative diseases in humans. Additionally, we outline the advantages and weaknesses of current in vitro and in vivo models and discuss future research directions investigating major human neurodegenerative diseases such as AD and Parkinson's diseases using dogs.
Collapse
Affiliation(s)
- Yoko M. Ambrosini
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Dana Borcherding
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Hyun Jung Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Auriel A. Willette
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Department of Food Science and Human Nutrition, College of Agriculture and Life Sciences, Iowa State University, Ames, IA, United States
| | - Albert Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
41
|
Role of iPSC-derived pericytes on barrier function of iPSC-derived brain microvascular endothelial cells in 2D and 3D. Fluids Barriers CNS 2019; 16:15. [PMID: 31167667 PMCID: PMC6551886 DOI: 10.1186/s12987-019-0136-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/08/2019] [Indexed: 01/25/2023] Open
Abstract
Background Pericytes of the blood–brain barrier (BBB) are embedded within basement membrane between brain microvascular endothelial cells (BMECs) and astrocyte end-feet. Despite the direct cell–cell contact observed in vivo, most in vitro BBB models introduce an artificial membrane that separates pericytes from BMECs. In this study, we investigated the effects of pericytes on BMEC barrier function across a range of in vitro platforms with varied spatial orientations and levels of cell–cell contact. Methods We differentiated RFP-pericytes and GFP-BMECs from hiPSCs and monitored transendothelial electrical resistance (TEER) across BMECs on transwell inserts while pericytes were either directly co-cultured on the membrane, indirectly co-cultured in the basolateral chamber, or embedded in a collagen I gel formed on the transwell membrane. We then incorporated pericytes into a tissue-engineered microvessel model of the BBB and measured pericyte motility and microvessel permeability. Results We found that BMEC monolayers did not require co-culture with pericytes to achieve physiological TEER values (> 1500 Ω cm2). However, under stressed conditions where TEER values for BMEC monolayers were reduced, indirectly co-cultured hiPSC-derived pericytes restored optimal TEER. Conversely, directly co-cultured pericytes resulted in a decrease in TEER by interfering with BMEC monolayer continuity. In the microvessel model, we observed direct pericyte-BMEC contact, abluminal pericyte localization, and physiologically-low Lucifer yellow permeability comparable to that of BMEC microvessels. In addition, pericyte motility decreased during the first 48 h of co-culture, suggesting progression towards pericyte stabilization. Conclusions We demonstrated that monocultured BMECs do not require co-culture to achieve physiological TEER, but that suboptimal TEER in stressed monolayers can be increased through co-culture with hiPSC-derived pericytes or conditioned media. We also developed the first BBB microvessel model using exclusively hiPSC-derived BMECs and pericytes, which could be used to examine vascular dysfunction in the human CNS. Electronic supplementary material The online version of this article (10.1186/s12987-019-0136-7) contains supplementary material, which is available to authorized users.
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW We review current knowledge regarding HDL and Alzheimer's disease, focusing on HDL's vasoprotective functions and potential as a biomarker and therapeutic target for the vascular contributions of Alzheimer's disease. RECENT FINDINGS Many epidemiological studies have observed that circulating HDL levels associate with decreased Alzheimer's disease risk. However, it is now understood that the functions of HDL may be more informative than levels of HDL cholesterol (HDL-C). Animal model studies demonstrate that HDL protects against memory deficits, neuroinflammation, and cerebral amyloid angiopathy (CAA). In-vitro studies using state-of-the-art 3D models of the human blood-brain barrier (BBB) confirm that HDL reduces vascular Aβ accumulation and attenuates Aβ-induced endothelial inflammation. Although HDL-based therapeutics have not been tested in clinical trials for Alzheimer's disease , several HDL formulations are in advanced phase clinical trials for coronary artery disease and atherosclerosis and could be leveraged toward Alzheimer's disease . SUMMARY Evidence from human studies, animal models, and bioengineered arteries supports the hypothesis that HDL protects against cerebrovascular dysfunction in Alzheimer's disease. Assays of HDL functions relevant to Alzheimer's disease may be desirable biomarkers of cerebrovascular health. HDL-based therapeutics may also be of interest for Alzheimer's disease, using stand-alone or combination therapy approaches.
Collapse
Affiliation(s)
- Emily B. Button
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jérôme Robert
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tara M. Caffrey
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jianjia Fan
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wenchen Zhao
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
43
|
Zhao H, Chappell JC. Microvascular bioengineering: a focus on pericytes. J Biol Eng 2019; 13:26. [PMID: 30984287 PMCID: PMC6444752 DOI: 10.1186/s13036-019-0158-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/15/2019] [Indexed: 12/26/2022] Open
Abstract
Capillaries within the microcirculation are essential for oxygen delivery and nutrient/waste exchange, among other critical functions. Microvascular bioengineering approaches have sought to recapitulate many key features of these capillary networks, with an increasing appreciation for the necessity of incorporating vascular pericytes. Here, we briefly review established and more recent insights into important aspects of pericyte identification and function within the microvasculature. We then consider the importance of including vascular pericytes in various bioengineered microvessel platforms including 3D culturing and microfluidic systems. We also discuss how vascular pericytes are a vital component in the construction of computational models that simulate microcirculation phenomena including angiogenesis, microvascular biomechanics, and kinetics of exchange across the vessel wall. In reviewing these topics, we highlight the notion that incorporating pericytes into microvascular bioengineering applications will increase their utility and accelerate the translation of basic discoveries to clinical solutions for vascular-related pathologies.
Collapse
Affiliation(s)
- Huaning Zhao
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA
| | - John C Chappell
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA.,3Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016 USA
| |
Collapse
|
44
|
Logan S, Arzua T, Canfield SG, Seminary ER, Sison SL, Ebert AD, Bai X. Studying Human Neurological Disorders Using Induced Pluripotent Stem Cells: From 2D Monolayer to 3D Organoid and Blood Brain Barrier Models. Compr Physiol 2019; 9:565-611. [PMID: 30873582 PMCID: PMC6705133 DOI: 10.1002/cphy.c180025] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurological disorders have emerged as a predominant healthcare concern in recent years due to their severe consequences on quality of life and prevalence throughout the world. Understanding the underlying mechanisms of these diseases and the interactions between different brain cell types is essential for the development of new therapeutics. Induced pluripotent stem cells (iPSCs) are invaluable tools for neurological disease modeling, as they have unlimited self-renewal and differentiation capacity. Mounting evidence shows: (i) various brain cells can be generated from iPSCs in two-dimensional (2D) monolayer cultures; and (ii) further advances in 3D culture systems have led to the differentiation of iPSCs into organoids with multiple brain cell types and specific brain regions. These 3D organoids have gained widespread attention as in vitro tools to recapitulate complex features of the brain, and (iii) complex interactions between iPSC-derived brain cell types can recapitulate physiological and pathological conditions of blood-brain barrier (BBB). As iPSCs can be generated from diverse patient populations, researchers have effectively applied 2D, 3D, and BBB models to recapitulate genetically complex neurological disorders and reveal novel insights into molecular and genetic mechanisms of neurological disorders. In this review, we describe recent progress in the generation of 2D, 3D, and BBB models from iPSCs and further discuss their limitations, advantages, and future ventures. This review also covers the current status of applications of 2D, 3D, and BBB models in drug screening, precision medicine, and modeling a wide range of neurological diseases (e.g., neurodegenerative diseases, neurodevelopmental disorders, brain injury, and neuropsychiatric disorders). © 2019 American Physiological Society. Compr Physiol 9:565-611, 2019.
Collapse
Affiliation(s)
- Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott G. Canfield
- Department of Cellular & Integrative Physiology, IU School of Medicine-Terre Haute, Terre Haute, IN, USA
| | - Emily R. Seminary
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samantha L. Sison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
45
|
Effect of flow on targeting and penetration of angiopep-decorated nanoparticles in a microfluidic model blood-brain barrier. PLoS One 2018; 13:e0205158. [PMID: 30300391 PMCID: PMC6177192 DOI: 10.1371/journal.pone.0205158] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 09/20/2018] [Indexed: 12/27/2022] Open
Abstract
The blood-brain barrier (BBB) limits transport of nanoparticles from the circulation to the brain parenchyma. Angiopep-2, a peptide which functions as a brain transport vector, can be coupled to nanoparticles in order to facilitate binding and internalization by brain endothelial cells (ECs), and subsequent BBB penetration. This multi-step process may be affected by blood flow over brain ECs, as flow influences endothelial cell phenotype as well as interactions of nanoparticles with ECs. In the present study a microfluidic BBB model was constructed to evaluate binding and internalization by brain ECs, as well as BBB penetration of Angiopep-2 coupled liposomes (Ang2-Liposomes) in static and flow conditions. Ang2 conjugation to liposomes markedly improved binding relative to unconjugated liposomes. Ang2-Liposomes bound and were internalized efficiently by brain endothelial cells after static incubation or with 1 dyne/cm2 of fluid shear stress (FSS), while binding was reduced at a FSS of 6 dyne/cm2. Penetration of the model microfluidic BBB by Ang2-Liposomes was higher at a FSS of 1 dyne/cm2 and 6 dyne/cm2 than with static incubation. Analysis of barrier function and control experiments for receptor-mediated penetration provided insight into the magnitude of transcellular versus paracellular transport at each tested FSS. Overall, the results demonstrate that flow impacted the binding and BBB penetration of Ang2-functionalized nanoparticles. This highlights the relevance of the local flow environment for in vitro modeling of the performance of nanoparticles functionalized with BBB penetrating ligands.
Collapse
|
46
|
Potjewyd G, Moxon S, Wang T, Domingos M, Hooper NM. Tissue Engineering 3D Neurovascular Units: A Biomaterials and Bioprinting Perspective. Trends Biotechnol 2018; 36:457-472. [DOI: 10.1016/j.tibtech.2018.01.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/14/2022]
|
47
|
Martins C, Araújo F, Gomes MJ, Fernandes C, Nunes R, Li W, Santos HA, Borges F, Sarmento B. Using microfluidic platforms to develop CNS-targeted polymeric nanoparticles for HIV therapy. Eur J Pharm Biopharm 2018; 138:111-124. [PMID: 29397261 DOI: 10.1016/j.ejpb.2018.01.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/26/2022]
Abstract
The human immunodeficiency virus (HIV) uses the brain as reservoir, which turns it as a promising target to fight this pathology. Nanoparticles (NPs) of poly(lactic-co-glycolic) acid (PLGA) are potential carriers of anti-HIV drugs to the brain, since most of these antiretrovirals, as efavirenz (EFV), cannot surpass the blood-brain barrier (BBB). Forasmuch as the conventional production methods lack precise control over the final properties of particles, microfluidics emerged as a prospective alternative. This study aimed at developing EFV-loaded PLGA NPs through a conventional and microfluidic method, targeted to the BBB, in order to treat HIV neuropathology. Compared to the conventional method, NPs produced through microfluidics presented reduced size (73 nm versus 133 nm), comparable polydispersity (around 0.090), less negative zeta-potential (-14.1 mV versus -28.0 mV), higher EFV association efficiency (80.7% versus 32.7%) and higher drug loading (10.8% versus 3.2%). The microfluidics-produced NPs also demonstrated a sustained in vitro EFV release (50% released within the first 24 h). NPs functionalization with a transferrin receptor-binding peptide, envisaging BBB targeting, proved to be effective concerning nuclear magnetic resonance analysis (δ = -0.008 ppm; δ = -0.017 ppm). NPs demonstrated to be safe to BBB endothelial and neuron cells (metabolic activity above 70%), as well as non-hemolytic (1-2% of hemolysis, no morphological alterations on erythrocytes). Finally, functionalized nanosystems were able to interact more efficiently with BBB cells, and permeability of EFV associated with NPs through a BBB in vitro model was around 1.3-fold higher than the free drug.
Collapse
Affiliation(s)
- Cláudia Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Francisca Araújo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria João Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Carlos Fernandes
- CIQUP - Centro de Investigação em Química, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Rute Nunes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Wei Li
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, FI-00014 Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, FI-00014 Helsinki, Finland; HiLIFE - Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland
| | - Fernanda Borges
- CIQUP - Centro de Investigação em Química, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal.
| |
Collapse
|
48
|
Pericytes Derived from Human Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1109:111-124. [DOI: 10.1007/978-3-030-02601-1_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|