1
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [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: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
2
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2025; 599:599-644. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [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] [Received: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
3
|
Vollmuth N, Sin J, Kim BJ. Host-microbe interactions at the blood-brain barrier through the lens of induced pluripotent stem cell-derived brain-like endothelial cells. mBio 2024; 15:e0286223. [PMID: 38193670 PMCID: PMC10865987 DOI: 10.1128/mbio.02862-23] [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: 01/10/2024] Open
Abstract
Microbe-induced meningoencephalitis/meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when pathogens are able to cross the blood-brain barrier (BBB) and gain access to the CNS. The BBB consists of highly specialized brain endothelial cells that exhibit specific properties to allow tight regulation of CNS homeostasis and prevent pathogen crossing. However, during meningoencephalitis/meningitis, the BBB fails to protect the CNS. Modeling the BBB remains a challenge due to the specialized characteristics of these cells. In this review, we cover the induced pluripotent stem cell-derived, brain-like endothelial cell model during host-pathogen interaction, highlighting the strengths and recent work on various pathogens known to interact with the BBB. As stem cell technologies are becoming more prominent, the stem cell-derived, brain-like endothelial cell model has been able to reveal new insights in vitro, which remain challenging with other in vitro cell-based models consisting of primary human brain endothelial cells and immortalized human brain endothelial cell lines.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
4
|
Markovich Z, Abreu A, Sheng Y, Han SM, Xiao R. Deciphering internal and external factors influencing intestinal junctional complexes. Gut Microbes 2024; 16:2389320. [PMID: 39150987 PMCID: PMC11332634 DOI: 10.1080/19490976.2024.2389320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/18/2024] Open
Abstract
The intestinal barrier, an indispensable guardian of gastrointestinal health, mediates the intricate exchange between internal and external environments. Anchored by evolutionarily conserved junctional complexes, this barrier meticulously regulates paracellular permeability in essentially all living organisms. Disruptions in intestinal junctional complexes, prevalent in inflammatory bowel diseases and irritable bowel syndrome, compromise barrier integrity and often lead to the notorious "leaky gut" syndrome. Critical to the maintenance of the intestinal barrier is a finely orchestrated network of intrinsic and extrinsic factors that modulate the expression, composition, and functionality of junctional complexes. This review navigates through the composition of key junctional complex components and the common methods used to assess intestinal permeability. It also explores the critical intracellular signaling pathways that modulate these junctional components. Lastly, we delve into the complex dynamics between the junctional complexes, microbial communities, and environmental chemicals in shaping the intestinal barrier function. Comprehending this intricate interplay holds paramount importance in unraveling the pathophysiology of gastrointestinal disorders. Furthermore, it lays the foundation for the development of precise therapeutic interventions targeting barrier dysfunction.
Collapse
Affiliation(s)
- Zachary Markovich
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Smell and Taste, University of Florida, Gainesville, FL, USA
| | - Adriana Abreu
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yi Sheng
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sung Min Han
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rui Xiao
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL, USA
- Center for Smell and Taste, University of Florida, Gainesville, FL, USA
- Institute on Aging, University of Florida, Gainesville, FL, USA
- Genetics Institute, University of Florida, Gainesville, FL, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| |
Collapse
|
5
|
Gonzales-Aloy E, Ahmed-Cox A, Tsoli M, Ziegler DS, Kavallaris M. From cells to organoids: The evolution of blood-brain barrier technology for modelling drug delivery in brain cancer. Adv Drug Deliv Rev 2023; 196:114777. [PMID: 36931346 DOI: 10.1016/j.addr.2023.114777] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Brain cancer remains the deadliest cancer. The blood-brain barrier (BBB) is impenetrable to most drugs and is a complex 3D network of multiple cell types including endothelial cells, astrocytes, and pericytes. In brain cancers, the BBB becomes disrupted during tumor progression and forms the blood-brain tumor barrier (BBTB). To advance therapeutic development, there is a critical need for physiologically relevant BBB in vitro models. 3D cell systems are emerging as valuable preclinical models to accelerate discoveries for diseases. Given the versatility and capability of 3D cell models, their potential for modelling the BBB and BBTB is reviewed. Technological advances of BBB models and challenges of in vitro modelling the BBTB, and application of these models as tools for assessing therapeutics and nano drug delivery, are discussed. Quantitative, in vitro BBB models that are predictive of effective brain cancer therapies will be invaluable for accelerating advancing new treatments to the clinic.
Collapse
Affiliation(s)
- Estrella Gonzales-Aloy
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Katharina Gaus Light Microscopy Facility, Mark Wainright Analytical Center, UNSW Sydney, NSW, Australia
| | - Maria Tsoli
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Kids Cancer Center, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; UNSW RNA Institute, UNSW Sydney, NSW, Australia.
| |
Collapse
|
6
|
Du F, Shusta EV, Palecek SP. Extracellular matrix proteins in construction and function of in vitro blood-brain barrier models. FRONTIERS IN CHEMICAL ENGINEERING 2023. [DOI: 10.3389/fceng.2023.1130127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
The blood-brain barrier (BBB) is a highly impermeable barrier separating circulating blood and brain tissue. A functional BBB is critical for brain health, and BBB dysfunction has been linked to the pathophysiology of diseases such as stroke and Alzheimer’s disease. A variety of models have been developed to study the formation and maintenance of the BBB, ranging from in vivo animal models to in vitro models consisting of primary cells or cells differentiated from human pluripotent stem cells (hPSCs). These models must consider the composition and source of the cellular components of the neurovascular unit (NVU), including brain microvascular endothelial cells (BMECs), brain pericytes, astrocytes, and neurons, and how these cell types interact. In addition, the non-cellular components of the BBB microenvironment, such as the brain vascular basement membrane (BM) that is in direct contact with the NVU, also play key roles in BBB function. Here, we review how extracellular matrix (ECM) proteins in the brain vascular BM affect the BBB, with a particular focus on studies using hPSC-derived in vitro BBB models, and discuss how future studies are needed to advance our understanding of how the ECM affects BBB models to improve model performance and expand our knowledge on the formation and maintenance of the BBB.
Collapse
|
7
|
van Schaik PEM, Zuhorn IS, Baron W. Targeting Fibronectin to Overcome Remyelination Failure in Multiple Sclerosis: The Need for Brain- and Lesion-Targeted Drug Delivery. Int J Mol Sci 2022; 23:8418. [PMID: 35955549 PMCID: PMC9368816 DOI: 10.3390/ijms23158418] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neuroinflammatory and neurodegenerative disease with unknown etiology that can be characterized by the presence of demyelinated lesions. Prevailing treatment protocols in MS rely on the modulation of the inflammatory process but do not impact disease progression. Remyelination is an essential factor for both axonal survival and functional neurological recovery but is often insufficient. The extracellular matrix protein fibronectin contributes to the inhibitory environment created in MS lesions and likely plays a causative role in remyelination failure. The presence of the blood-brain barrier (BBB) hinders the delivery of remyelination therapeutics to lesions. Therefore, therapeutic interventions to normalize the pathogenic MS lesion environment need to be able to cross the BBB. In this review, we outline the multifaceted roles of fibronectin in MS pathogenesis and discuss promising therapeutic targets and agents to overcome fibronectin-mediated inhibition of remyelination. In addition, to pave the way for clinical use, we reflect on opportunities to deliver MS therapeutics to lesions through the utilization of nanomedicine and discuss strategies to deliver fibronectin-directed therapeutics across the BBB. The use of well-designed nanocarriers with appropriate surface functionalization to cross the BBB and target the lesion sites is recommended.
Collapse
Affiliation(s)
- Pauline E. M. van Schaik
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| | - Inge S. Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wia Baron
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands;
| |
Collapse
|
8
|
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: 18] [Impact Index Per Article: 6.0] [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
|
9
|
Abstract
The human blood-brain-barrier (BBB) is a vital structure for brain health. Conversely it represents a challenge in drug development programmes that require breaching of the barrier in order to access the central nervous system. Very often brain disorders have early dysfunction of the BBB implicating an important role in pathogenesis and disease progression. The development of human in vitro models is a major advance to allow experimental studies and screening assays, although there remain outstanding questions for the field. In this chapter, the current state of the art will be reviewed, with the complementary innovative approaches to in vitro modelling described, from simple 2D-cultures to more complex multi-cell type micro-physiological systems.
Collapse
Affiliation(s)
- Zameel Cader
- Translational Molecular Neuroscience Group, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Kurmann L, Okoniewski M, Dubey RK. Transcryptomic Analysis of Human Brain -Microvascular Endothelial Cell Driven Changes in -Vascular Pericytes. Cells 2021; 10:cells10071784. [PMID: 34359953 PMCID: PMC8304094 DOI: 10.3390/cells10071784] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Many pathological conditions of the brain are associated with structural abnormalities within the neurovascular system and linked to pericyte (PC) loss and/or dysfunction. Since crosstalk between endothelial cells (ECs) and PCs greatly impacts the function of the blood–brain barrier (BBB), effects of PCs on endothelial integrity and function have been investigated extensively. However, the impact of ECs on the function and activity of PCs remains largely unknown. Hence, using co-cultures of human brain vascular PCs with human cerebral microvascular ECs on opposite sides of porous Transwell inserts which facilitates direct EC–PC contact and improves EC barrier function, we analyzed EC-driven transcriptomic changes in PCs using microarrays and changes in cytokines/chemokines using proteome arrays. Gene expression analysis (GEA) in PCs co-cultured with ECs versus PCs cultured alone showed significant upregulation of 1′334 genes and downregulation of 964 genes. GEA in co-cultured PCs revealed increased expression of five prominent PC markers as well as soluble factors, such as transforming growth factor beta, fibroblast growth factor, angiopoietin 1, brain-derived neurotrophic factor, all of which are involved in EC–PC crosstalk and BBB induction. Pathway enrichment analysis of modulated genes showed a strong impact on many inflammatory and extracellular matrix (ECM) pathways including interferon and interleukin signaling, TGF-β and interleukin-1 regulation of ECM, as well as on the mRNA processing pathway. Interestingly, while co-culture induced the mRNA expression of many chemokines and cytokines, including several CCL- and CXC-motif ligands and interleukins, we observed a decreased expression of the same inflammatory mediators on the protein level. Importantly, in PCs, ECs significantly induced interferon associated proteins (IFIT1, IFI44L, IF127, IFIT3, IFI6, IFI44) with anti-viral actions; downregulated prostaglandin E receptor 2 (prevent COX-2 mediated BBB damage); upregulated fibulin-3 and connective tissue growth factor essential for BBB integrity; and multiple ECMs (collagens and integrins) that inhibit cell migration. Our findings suggest that via direct contact, ECs prime PCs to induce molecules to promote BBB integrity and cell survival during infection and inflammatory insult. Taken together, we provide first evidence that interaction with ECs though porous membranes induces major changes in the transcriptomic and proteomic profile of PCs. ECs influence genes involved in diverse aspects of PC function including PC maturation, cell survival, anti-viral defense, blood flow regulation, immuno-modulation and ECM deposition.
Collapse
Affiliation(s)
- Lisa Kurmann
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland;
| | | | - Raghvendra K. Dubey
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland;
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Correspondence:
| |
Collapse
|
11
|
Lasp1 regulates adherens junction dynamics and fibroblast transformation in destructive arthritis. Nat Commun 2021; 12:3624. [PMID: 34131132 PMCID: PMC8206096 DOI: 10.1038/s41467-021-23706-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 05/11/2021] [Indexed: 02/05/2023] Open
Abstract
The LIM and SH3 domain protein 1 (Lasp1) was originally cloned from metastatic breast cancer and characterised as an adaptor molecule associated with tumourigenesis and cancer cell invasion. However, the regulation of Lasp1 and its function in the aggressive transformation of cells is unclear. Here we use integrative epigenomic profiling of invasive fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA) and from mouse models of the disease, to identify Lasp1 as an epigenomically co-modified region in chronic inflammatory arthritis and a functionally important binding partner of the Cadherin-11/β-Catenin complex in zipper-like cell-to-cell contacts. In vitro, loss or blocking of Lasp1 alters pathological tissue formation, migratory behaviour and platelet-derived growth factor response of arthritic FLS. In arthritic human TNF transgenic mice, deletion of Lasp1 reduces arthritic joint destruction. Therefore, we show a function of Lasp1 in cellular junction formation and inflammatory tissue remodelling and identify Lasp1 as a potential target for treating inflammatory joint disorders associated with aggressive cellular transformation.
Collapse
|
12
|
Fessel J. Does synaptic hypometabolism or synaptic dysfunction, originate cognitive loss? Analysis of the evidence. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12177. [PMID: 34027027 PMCID: PMC8129845 DOI: 10.1002/trc2.12177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/31/2021] [Indexed: 11/25/2022]
Abstract
Elderly persons with currently normal cognition who have cerebral hypometabolism as shown by low uptake of 18fluorine-fluorodeoxyglucose (18F-FDG), are at risk of future loss of cognition and, thus, of future Alzheimer's dementia (AD). Reduction of either 18F-FDG or cognition is assumed to reflect synaptic dysfunction, since synapses account for the majority of glucose use by the brain and cognition depends upon accurate synaptic function. The chronology of the connection between reduced cerebral synaptic function and hypometabolism is, therefore, a critical question, because if synaptic dysfunction came first, then correcting the hypometabolism would likely not benefit synaptic function; but if hypometabolism came first, then correcting the hypometabolism probably would benefit synaptic function. That correction might prevent initiation of the cognitive loss that eventuates in AD and, thereby, would benefit the vast numbers of persons in their eighth to tenth decades of life who are at risk for AD. Among the many citations reviewed in this presentation, seven show hypometabolism that precedes synaptic dysfunction, and two show the reverse. Thus the preponderance of evidence, 78%, suggests that the initiating event is synaptic hypometabolism and that it is 3.5-fold less likely that synaptic dysfunction is the initiator. In addition, it is inherently unlikely that synaptic dysfunction causes hypometabolism. This conclusion could be tested by a clinical trial whose primary objective would be to assess the benefit to cognition of improving synaptic metabolism in patients who are at risk for cognitive loss.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
| |
Collapse
|
13
|
Kurmann L, Okoniewski M, Ogunshola OO, Leeners B, Imthurn B, Dubey RK. Transcryptomic Analysis of Human Brain-Microvascular Endothelial Response to -Pericytes: Cell Orientation Defines Barrier Function. Cells 2021; 10:cells10040963. [PMID: 33924251 PMCID: PMC8074760 DOI: 10.3390/cells10040963] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Pericytes facilitate blood-brain barrier (BBB) integrity; however, the mechanisms involved remain unclear. Hence, using co-cultures of human cerebral microvascular endothelial cells (ECs) and vascular pericytes (PCs) in different spatial arrangements, as well as PC conditioned media, we investigated the impact of PC-EC orientation and PC-derived soluble factors on EC barrier function. We provide the first evidence that barrier-inducing properties of PCs require basolateral contact with ECs. Gene expression analysis (GEA) in ECs co-cultured with PCs versus ECs alone showed significant upregulation of 38 genes and downregulation of 122 genes. Pathway enrichment analysis of modulated genes showed significant regulation of several pathways, including transforming growth factor-β and interleukin-1 regulated extracellular matrix, interferon and interleukin signaling, immune system signaling, receptor of advanced glycation end products (RAGE), and cytokine-cytokine receptor interaction. Transcriptomic analysis showed a reduction in molecules such as pro-inflammatory cytokines and chemokines, which are known to be induced during BBB disruption. Moreover, cytokine proteome array confirmed the downregulation of key pro-inflammatory cytokines and chemokines on the protein level. Other molecules which influence BBB and were favorably modulated upon EC-PC co-culture include IL-18 binding protein, kallikrein-3, CSF2 CSF3, CXCL10, CXCL11 (downregulated) and IL-1-R4; HGF, PDGF-AB/BB, PECAM, SERPIN E1 (upregulated). In conclusion, we provide the first evidence that (1) basolateral contact between ECs and PCs is essential for EC barrier function and integrity; (2) in ECs co-cultured with PCs, the profile of BBB disrupting pro-inflammatory molecules and cytokines/chemokines is downregulated; (3) PCs significantly modulate EC mechanisms known to improve barrier function, including TGF-β regulated ECM pathway, anti-inflammatory cytokines, growth factors and matrix proteins. This human PC-EC co-culture may serve as a viable in vitro model for investigating BBB function and drug transport.
Collapse
Affiliation(s)
- Lisa Kurmann
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland; (L.K.); (B.L.); (B.I.)
| | | | - Omolara O. Ogunshola
- Zurich Center Integrative Physiology (ZIHP), Vetsuisse Faculty, Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland;
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland; (L.K.); (B.L.); (B.I.)
| | - Bruno Imthurn
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland; (L.K.); (B.L.); (B.I.)
| | - Raghvendra K. Dubey
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland; (L.K.); (B.L.); (B.I.)
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Correspondence:
| |
Collapse
|
14
|
Stanković T, Ranđelović T, Dragoj M, Stojković Burić S, Fernández L, Ochoa I, Pérez-García VM, Pešić M. In vitro biomimetic models for glioblastoma-a promising tool for drug response studies. Drug Resist Updat 2021; 55:100753. [PMID: 33667959 DOI: 10.1016/j.drup.2021.100753] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
The poor response of glioblastoma to current treatment protocols is a consequence of its intrinsic drug resistance. Resistance to chemotherapy is primarily associated with considerable cellular heterogeneity, and plasticity of glioblastoma cells, alterations in gene expression, presence of specific tumor microenvironment conditions and blood-brain barrier. In an attempt to successfully overcome chemoresistance and better understand the biological behavior of glioblastoma, numerous tri-dimensional (3D) biomimetic models were developed in the past decade. These novel advanced models are able to better recapitulate the spatial organization of glioblastoma in a real time, therefore providing more realistic and reliable evidence to the response of glioblastoma to therapy. Moreover, these models enable the fine-tuning of different tumor microenvironment conditions and facilitate studies on the effects of the tumor microenvironment on glioblastoma chemoresistance. This review outlines current knowledge on the essence of glioblastoma chemoresistance and describes the progress achieved by 3D biomimetic models. Moreover, comprehensive literature assessment regarding the influence of 3D culturing and microenvironment mimicking on glioblastoma gene expression and biological behavior is also provided. The contribution of the blood-brain barrier as well as the blood-tumor barrier to glioblastoma chemoresistance is also reviewed from the perspective of 3D biomimetic models. Finally, the role of mathematical models in predicting 3D glioblastoma behavior and drug response is elaborated. In the future, technological innovations along with mathematical simulations should create reliable 3D biomimetic systems for glioblastoma research that should facilitate the identification and possibly application in preclinical drug testing and precision medicine.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Teodora Ranđelović
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Miodrag Dragoj
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Luis Fernández
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Victor M Pérez-García
- Departamento de Matemáticas, E.T.S.I. Industriales and Instituto de Matemática Aplicada a la Ciencia y la Ingeniería (IMACI), Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia.
| |
Collapse
|
15
|
Towards the development of a human in vitro model of the blood-brain barrier for virus-associated acute encephalopathy: assessment of the time- and concentration-dependent effects of TNF-α on paracellular tightness. Exp Brain Res 2020; 239:451-461. [PMID: 33219841 DOI: 10.1007/s00221-020-05985-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/09/2020] [Indexed: 12/30/2022]
Abstract
The pathogenesis of virus-associated acute encephalopathy (VAE) involves brain edema caused by disruption of the blood-brain barrier (BBB). We aimed to develop an in vitro VAE model using an in vitro BBB model, to evaluate the dynamics of vascular dysfunction caused by tumor necrosis factor (TNF)-α. A co-culture model, consisting of Transwell®-grown human brain microvascular endothelial cells and pericytes, was treated with serially diluted TNF-α. Transendothelial electrical resistance (TER) was measured using cellZscope®. A permeability assay, using fluorescein isothiocyanate-conjugated sodium or dextran, was performed. Changes in claudin-5 localization and expression after TNF-α treatment were observed using immunofluorescence staining and western blot analysis. The TER decreased and permeability increased after TNF-α treatment; recovery time was dependent on TNF-α concentration. Claudin-5 was delocalized after TNF-α treatment and recovered in a TNF-α concentration-dependent manner. The expression of claudin-5 decreased 24 h after the TNF-α treatment and completely recovered 48 h after TNF-α treatment. Claudin-5 delocalization was likely associated with vascular hyperpermeability. To conclude, we evaluated vascular endothelial cell permeability and injury in VAE using an in vitro BBB model treated with TNF-α. This system can be useful for developing novel therapeutic strategies for VAE and designing treatments that target vascular permeability.
Collapse
|
16
|
Morofuji Y, Nakagawa S. Drug Development for Central Nervous System Diseases Using In vitro Blood-brain Barrier Models and Drug Repositioning. Curr Pharm Des 2020; 26:1466-1485. [PMID: 32091330 PMCID: PMC7499354 DOI: 10.2174/1381612826666200224112534] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/30/2020] [Indexed: 12/15/2022]
Abstract
An important goal of biomedical research is to translate basic research findings into practical clinical implementation. Despite the advances in the technology used in drug discovery, the development of drugs for central nervous system diseases remains challenging. The failure rate for new drugs targeting important central nervous system diseases is high compared to most other areas of drug discovery. The main reason for the failure is the poor penetration efficacy across the blood-brain barrier. The blood-brain barrier represents the bottleneck in central nervous system drug development and is the most important factor limiting the future growth of neurotherapeutics. Meanwhile, drug repositioning has been becoming increasingly popular and it seems a promising field in central nervous system drug development. In vitro blood-brain barrier models with high predictability are expected for drug development and drug repositioning. In this review, the recent progress of in vitro BBB models and the drug repositioning for central nervous system diseases will be discussed.
Collapse
Affiliation(s)
- Yoichi Morofuji
- Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8501, Japan
| | - Shinsuke Nakagawa
- Department of Medical Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
17
|
Abasi S, Bhat A, Guiseppi‐Elie A. Electrode Selection for Electrostimulation and TEER Using ECSARA. ELECTROANAL 2020. [DOI: 10.1002/elan.202060313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Sara Abasi
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
| | - Ankita Bhat
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
| | - Anthony Guiseppi‐Elie
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
- Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute 6670 Bertner Ave. Houston TX 77030 USA
- Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
- ABTECH Scientific, Inc. Biotechnology Research Park 800 East Leigh Street Richmond VA 23219 USA
| |
Collapse
|
18
|
Salman MM, Marsh G, Kusters I, Delincé M, Di Caprio G, Upadhyayula S, de Nola G, Hunt R, Ohashi KG, Gray T, Shimizu F, Sano Y, Kanda T, Obermeier B, Kirchhausen T. Design and Validation of a Human Brain Endothelial Microvessel-on-a-Chip Open Microfluidic Model Enabling Advanced Optical Imaging. Front Bioeng Biotechnol 2020; 8:573775. [PMID: 33117784 PMCID: PMC7576009 DOI: 10.3389/fbioe.2020.573775] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/21/2020] [Indexed: 01/30/2023] Open
Abstract
We describe here the design and implementation of an in vitro microvascular open model system using human brain microvascular endothelial cells. The design has several advantages over other traditional closed microfluidic platforms: (1) it enables controlled unidirectional flow of media at physiological rates to support vascular function, (2) it allows for very small volumes which makes the device ideal for studies involving biotherapeutics, (3) it is amenable for multiple high resolution imaging modalities such as transmission electron microscopy (TEM), 3D live fluorescence imaging using traditional spinning disk confocal microscopy, and advanced lattice light sheet microscopy (LLSM). Importantly, we miniaturized the design, so it can fit within the physical constraints of LLSM, with the objective to study physiology in live cells at subcellular level. We validated barrier function of our brain microvessel-on-a-chip by measuring permeability of fluorescent dextran and a human monoclonal antibody. One potential application is to investigate mechanisms of transcytosis across the brain microvessel-like barrier of fluorescently-tagged biologics, viruses or nanoparticles.
Collapse
Affiliation(s)
- Mootaz M. Salman
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | | | - Ilja Kusters
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Matthieu Delincé
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Giuseppe Di Caprio
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Srigokul Upadhyayula
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Giovanni de Nola
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Ronan Hunt
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | - Kazuka G. Ohashi
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
| | | | | | - Yasuteru Sano
- Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Kanda
- Yamaguchi University Graduate School of Medicine, Ube, Japan
| | | | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
19
|
Roldán DB, Grimmler M, Hartmann C, Hubich-Rau S, Beißert T, Paret C, Cagna G, Rohde C, Wöll S, Koslowski M, Türeci Ö, Sahin U. PLAC1 is essential for FGF7/FGFRIIIb-induced Akt-mediated cancer cell proliferation. Oncotarget 2020; 11:1862-1875. [PMID: 32499871 PMCID: PMC7244013 DOI: 10.18632/oncotarget.27582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/14/2020] [Indexed: 12/20/2022] Open
Abstract
PLAC1 (placenta enriched 1) is a mammalian trophoblast-specific protein. Aberrant expression of PLAC1 is observed in various human cancers, where it is involved in the motility, migration, and invasion of tumor cells, which are associated with the phosphoinositide 3-kinase (PI3K)/AKT pathway. We previously demonstrated that AKT activation mediates the downstream effects of PLAC1; however, the molecular mechanisms of PLAC1-induced AKT-mediated tumor-related processes are unclear. We studied human choriocarcinoma and breast cancer cell lines to explore the localization and receptor-ligand interactions, as well as the downstream effects of PLAC1. We show secretion and adherence of PLAC1 to the extracellular matrix, where it forms a trimeric complex with fibroblast growth factor 7 (FGF7) and its receptor, FGF receptor 2 IIIb (FGFR2IIIb). We further show that PLAC1 signaling via FGFR2IIIb activates AKT phosphorylation in cancer cell lines. As the FGF pathway is of major interest in anticancer therapeutic strategies, these data further promote PLAC1 as a promising anticancer drug target.
Collapse
Affiliation(s)
- Diana Barea Roldán
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Matthias Grimmler
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: DiaSys Diagnostic Systems GmbH, Holzheim, Germany
- These authors contributed equally to this work
| | - Christoph Hartmann
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
- These authors contributed equally to this work
| | - Stefanie Hubich-Rau
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- These authors contributed equally to this work
| | - Tim Beißert
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Claudia Paret
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Giuseppe Cagna
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Lonza Pharma & Biotech, Cologne, Germany
| | - Christoph Rohde
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Current address: Merck KGaA, Darmstadt, Germany
| | - Stefan Wöll
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Michael Koslowski
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Formerly of University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Current address: GammaDelta Therapeutics, London, United Kingdom
| | - Özlem Türeci
- Formerly of TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Formerly of Ganymed Pharmaceuticals AG, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- Ci3 Cluster for Individualized Immune Intervention, Mainz, Germany
| | - Ugur Sahin
- TRON–Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
20
|
Gray KM, Jung JW, Inglut CT, Huang HC, Stroka KM. Quantitatively relating brain endothelial cell-cell junction phenotype to global and local barrier properties under varied culture conditions via the Junction Analyzer Program. Fluids Barriers CNS 2020; 17:16. [PMID: 32046757 PMCID: PMC7014765 DOI: 10.1186/s12987-020-0177-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/31/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The endothelial cell-cell junctions of the blood-brain barrier (BBB) play a pivotal role in the barrier's function. Altered cell-cell junctions can lead to barrier dysfunction and have been implicated in several diseases. Despite this, the driving forces regulating junctional protein presentation remain relatively understudied, largely due to the lack of efficient techniques to quantify their presentation at sites of cell-cell adhesion. Here, we used our novel Junction Analyzer Program (JAnaP) to quantify junction phenotype (i.e., continuous, punctate, or perpendicular) in response to various substrate compositions, cell culture times, and cAMP treatments in human brain microvascular endothelial cells (HBMECs). We then quantitatively correlated junction presentation with barrier permeability on both a "global" and "local" scale. METHODS We cultured HBMECs on collagen I, fibronectin, collagen IV, laminin, fibronectin/collagen IV/laminin, or hyaluronic acid/gelatin for 2, 4, and 7 days with varying cAMP treatment schedules. Images of immunostained ZO-1, VE-cadherin, and claudin-5 were analyzed using the JAnaP to calculate the percent of the cell perimeter presenting continuous, punctate, or perpendicular junctions. Transwell permeability assays and resistance measurements were used to measure bulk ("global") barrier properties, and a "local" permeability assay was used to correlate junction presentation proximal to permeable monolayer regions. RESULTS Substrate composition was found to play little role in junction presentation, while cAMP supplements significantly increased the continuous junction architecture. Increased culture time required increased cAMP treatment time to reach similar ZO-1 and VE-cadherin coverage observed with shorter culture, though longer cultures were required for claudin-5 presentation. Prolonged cAMP treatment (6 days) disrupted junction integrity for all three junction proteins. Transwell permeability and TEER assays showed no correlation with junction phenotype, but a local permeability assay revealed a correlation between the number of discontinuous and no junction regions with barrier penetration. CONCLUSIONS These results suggest that cAMP signaling influences HBMEC junction architecture more than matrix composition. Our studies emphasized the need for local barrier measurement to mechanistically understand the role of junction phenotype and supported previous results that continuous junctions are indicative of a more mature/stable endothelial barrier. Understanding what conditions influence junction presentations, and how they, in turn, affect barrier integrity, could lead to the development of therapeutics for diseases associated with BBB dysfunction.
Collapse
Affiliation(s)
- Kelsey M Gray
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, 3110 A. James Clark Hall, College Park, MD, 20742, USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, 3110 A. James Clark Hall, College Park, MD, 20742, USA
| | - Collin T Inglut
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, 3110 A. James Clark Hall, College Park, MD, 20742, USA
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, 3110 A. James Clark Hall, College Park, MD, 20742, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland-Baltimore, Baltimore, MD, 21201, USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Drive, 3110 A. James Clark Hall, 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, Baltimore, MD, 21201, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland-Baltimore, Baltimore, MD, 21201, USA.
| |
Collapse
|
21
|
Gomez-Zepeda D, Taghi M, Scherrmann JM, Decleves X, Menet MC. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019; 12:pharmaceutics12010020. [PMID: 31878061 PMCID: PMC7022905 DOI: 10.3390/pharmaceutics12010020] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Drug delivery into the brain is regulated by the blood-brain interfaces. The blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the blood-arachnoid barrier (BAB) regulate the exchange of substances between the blood and brain parenchyma. These selective barriers present a high impermeability to most substances, with the selective transport of nutrients and transporters preventing the entry and accumulation of possibly toxic molecules, comprising many therapeutic drugs. Transporters of the ATP-binding cassette (ABC) superfamily have an important role in drug delivery, because they extrude a broad molecular diversity of xenobiotics, including several anticancer drugs, preventing their entry into the brain. Gliomas are the most common primary tumors diagnosed in adults, which are often characterized by a poor prognosis, notably in the case of high-grade gliomas. Therapeutic treatments frequently fail due to the difficulty of delivering drugs through the brain barriers, adding to diverse mechanisms developed by the cancer, including the overexpression or expression de novo of ABC transporters in tumoral cells and/or in the endothelial cells forming the blood-brain tumor barrier (BBTB). Many models have been developed to study the phenotype, molecular characteristics, and function of the blood-brain interfaces as well as to evaluate drug permeability into the brain. These include in vitro, in vivo, and in silico models, which together can help us to better understand their implication in drug resistance and to develop new therapeutics or delivery strategies to improve the treatment of pathologies of the central nervous system (CNS). In this review, we present the principal characteristics of the blood-brain interfaces; then, we focus on the ABC transporters present on them and their implication in drug delivery; next, we present some of the most important models used for the study of drug transport; finally, we summarize the implication of ABC transporters in glioma and the BBTB in drug resistance and the strategies to improve the delivery of CNS anticancer drugs.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| | - Méryam Taghi
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Xavier Decleves
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Biologie du médicament et toxicologie, Hôpital Cochin, AP HP, 75006 Paris, France
| | - Marie-Claude Menet
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Hormonologie adulte, Hôpital Cochin, AP HP, 75006 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| |
Collapse
|
22
|
Praça C, Rosa SC, Sevin E, Cecchelli R, Dehouck MP, Ferreira LS. Derivation of Brain Capillary-like Endothelial Cells from Human Pluripotent Stem Cell-Derived Endothelial Progenitor Cells. Stem Cell Reports 2019; 13:599-611. [PMID: 31495714 PMCID: PMC6829749 DOI: 10.1016/j.stemcr.2019.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
The derivation of human brain capillary endothelial cells is of utmost importance for drug discovery programs focusing on diseases of the central nervous system. Here, we describe a two-step differentiation protocol to derive brain capillary-like endothelial cells from human pluripotent stem cells. The cells were initially differentiated into endothelial progenitor cells followed by specification into a brain capillary-like endothelial cell phenotype using a protocol that combined the induction, in a time-dependent manner, of VEGF, Wnt3a, and retinoic acid signaling pathways and the use of fibronectin as the extracellular matrix. The brain capillary-like endothelial cells displayed a permeability to lucifer yellow of 1 × 10−3 cm/min, a transendothelial electrical resistance value of 60 Ω cm2 and were able to generate a continuous monolayer of cells expressing ZO-1 and CLAUDIN-5 but moderate expression of P-glycoprotein. Further maturation of these cells required coculture with pericytes. The study presented here opens a new approach for the study of soluble and non-soluble factors in the specification of endothelial progenitor cells into brain capillary-like endothelial cells. Derivation of BCLECs from iPSCs in chemically defined medium in two steps Specification of EPCs into BCLECs requires the activation of VEGF, Wnt3a, and RA Fibronectin seems a sufficient substrate for the specification of EPCs into BCLECs
Collapse
Affiliation(s)
- Catarina Praça
- Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Susana C Rosa
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Emmanuel Sevin
- Faculté des Sciences Jean Perrin, Université d'Artois, Lens, France
| | - Romeo Cecchelli
- Faculté des Sciences Jean Perrin, Université d'Artois, Lens, France
| | | | - Lino S Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
23
|
Zhou W, Graham K, Lucendo-Villarin B, Flint O, Hay DC, Bagnaninchi P. Combining stem cell-derived hepatocytes with impedance sensing to better predict human drug toxicity. Expert Opin Drug Metab Toxicol 2018; 15:77-83. [PMID: 30572740 DOI: 10.1080/17425255.2019.1558208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: The liver plays a central role in human drug metabolism. To model drug metabolism, the major cell type of the liver, the hepatocyte, is commonly used. Hepatocytes can be derived from human and animal sources, including pluripotent stem cells. Cell-based models have shown promise in modeling human drug exposure. The assays used in those studies are normally 'snap-shot' in nature, and do not provide the complete picture of human drug exposure. Research design and methods: In this study, we employ stem cell-derived hepatocytes and impedance sensing to model human drug toxicity. This impedance-based stem cell assay reports hepatotoxicity in real time after treatment with compounds provided by industry. Results: Using electric cell-substrate impedance Sensing (ECIS), we were able to accurately measure drug toxicity post-drug exposure in real time and more quickly than gold standard biochemical assays. Conclusions: ECIS is robust and non-destructive methodology capable of monitoring human drug exposure with superior performance to current gold standard 'snapshot' assays. We believe that the methodology presented within this article could prove valuable in the quest to better predict off-target effects of drugs in humans.
Collapse
Affiliation(s)
- Wenli Zhou
- a Department of Medical Oncology , Changzheng Hospital, Navy medical University , Shanghai , China
| | - Karen Graham
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - Baltasar Lucendo-Villarin
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - Oliver Flint
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - David C Hay
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| | - Pierre Bagnaninchi
- b MRC Centre for Regenerative Medicine, 5 Little France Drive , University of Edinburgh , Edinburgh , UK
| |
Collapse
|
24
|
In Vitro Cell Models of the Human Blood-Brain Barrier: Demonstrating the Beneficial Influence of Shear Stress on Brain Microvascular Endothelial Cell Phenotype. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-1-4939-8946-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
DeStefano JG, Jamieson JJ, Linville RM, Searson PC. Benchmarking in vitro tissue-engineered blood-brain barrier models. Fluids Barriers CNS 2018; 15:32. [PMID: 30514389 PMCID: PMC6280508 DOI: 10.1186/s12987-018-0117-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022] Open
Abstract
The blood–brain barrier (BBB) plays a key role in regulating transport into and out of the brain. With increasing interest in the role of the BBB in health and disease, there have been significant advances in the development of in vitro models. The value of these models to the research community is critically dependent on recapitulating characteristics of the BBB in humans or animal models. However, benchmarking in vitro models is surprisingly difficult since much of our knowledge of the structure and function of the BBB comes from in vitro studies. Here we describe a set of parameters that we consider a starting point for benchmarking and validation. These parameters are associated with structure (ultrastructure, wall shear stress, geometry), microenvironment (basement membrane and extracellular matrix), barrier function (transendothelial electrical resistance, permeability, efflux transport), cell function (expression of BBB markers, turnover), and co-culture with other cell types (astrocytes and pericytes). In suggesting benchmarks, we rely primarily on imaging or direct measurements in humans and animal models.
Collapse
Affiliation(s)
- Jackson G DeStefano
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John J Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA. .,120 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
26
|
Heidari H, Taylor H. Review Article: Capturing the physiological complexity of the brain's neuro-vascular unit in vitro. BIOMICROFLUIDICS 2018; 12:051502. [PMID: 30364144 PMCID: PMC6191301 DOI: 10.1063/1.5045126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 09/27/2018] [Indexed: 06/01/2023]
Abstract
With the accelerating pace of brain research in recent years and the growing appreciation of the complexity of the brain and several brain-associated neurological diseases, the demand for powerful tools to enhance drug screening, diagnosis, and fundamental research is greater than ever. Highly representative models of the central nervous system (CNS) can play a critical role in meeting these needs. Unfortunately, in vivo animal models lack controllability, are difficult to monitor, and do not model human-specific brain behavior accurately. On the other hand, in silico computational models struggle to capture comprehensively the intertwined biological, chemical, electrical, and mechanical complexity of the brain. This leaves us with the promising domain of "organ-on-chip" in vitro models. In this review, we describe some of the most pioneering efforts in this expanding field, offering a perspective on the new possibilities as well as the limitations of each approach. We focus particularly on how the models reproduce the blood-brain barrier (BBB), which mediates mass transport to and from brain tissue. We also offer a brief commentary on strategies for evaluating the blood-brain barrier functionality of these in vitro models, including trans-endothelial electrical resistance (TEER), immunocytochemistry, and permeability analysis. From the early membrane-based models of the BBB that have grown into the Transwell® class of devices, to the era of microfluidic chips and a future of bio-printed tissue, we see enormous improvement in the reliability of in vitro models. More and more of the biological and structural complexity of the BBB is being captured by microfluidic chips, and the organ-specificity of bio-printed tissue is also significantly improved. Although we believe that the long-term solution will eventually take the form of automated and parallelized bio-printing systems, we find that valuable transport studies can already be accomplished with microfluidic platforms.
Collapse
Affiliation(s)
- Hossein Heidari
- Department of Mechanical Engineering, University of California, 6159 Etcheverry Hall, Berkeley, California 94720, USA
| | - Hayden Taylor
- Department of Mechanical Engineering, University of California, 6159 Etcheverry Hall, Berkeley, California 94720, USA
| |
Collapse
|
27
|
Galla HJ. Monocultures of primary porcine brain capillary endothelial cells: Still a functional in vitro model for the blood-brain-barrier. J Control Release 2018; 285:172-177. [PMID: 30005905 DOI: 10.1016/j.jconrel.2018.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022]
Abstract
The main obstacle for the treatment of brain diseases is the restriction of the passage of pharmaceuticals across the blood-brain barrier. Endothelial cells line up the cerebral micro vessels and prevent the uncontrolled transfer of polar substances by intercellular tight junctions. In addition to this physical barrier, active transporters of the multi-drug-resistance prevent the passage of hydrophobic substances by refluxing them back to the blood stream. This paper reviews the development and selected applications of an in vitro porcine brain derived primary cell culture system established in the authors lab that closely resembles the BBB in vivo and could thus be used to study beyond other applications drug delivery to the brain. An essential technique to control the intactness or destruction of the barrier, the impedance spectroscopy, will be introduced. It will be shown that nanoparticles can cross the blood brain barrier by two mechanisms: opening the tight junctions and thus allowing parallel import of substances into the brain as well as receptor mediated endocytosis using brain specific target molecules. However cytotoxic effects have to be considered as well which beside standard cytotoxicity assays could be also determined by impedance technology. Moreover it will be shown that enzymes e.g. for enzyme replacement therapy could be transferred across the barrier by proper tuning or chemical modification of the enzyme. Since this review is based on a conference presentation it will mainly focus on applications of the monoculture system developed in the authors lab which under given culture conditions is useful due to its easy availability, robustness, good reproducibility and also due to its simplicity. Improvements of this model made by other groups will be acknowledged but not discussed here in detail.
Collapse
Affiliation(s)
- Hans-Joachim Galla
- Institute for Biochemistry, Westfälische Wilhems Universität,Münster, Wilhelm Klemm Str. 2, 48149 Münster, Germany.
| |
Collapse
|
28
|
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: 8.6] [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]
|
29
|
Modarres HP, Janmaleki M, Novin M, Saliba J, El-Hajj F, RezayatiCharan M, Seyfoori A, Sadabadi H, Vandal M, Nguyen MD, Hasan A, Sanati-Nezhad A. In vitro models and systems for evaluating the dynamics of drug delivery to the healthy and diseased brain. J Control Release 2018; 273:108-130. [PMID: 29378233 DOI: 10.1016/j.jconrel.2018.01.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/12/2022]
Abstract
The blood-brain barrier (BBB) plays a crucial role in maintaining brain homeostasis and transport of drugs to the brain. The conventional animal and Transwell BBB models along with emerging microfluidic-based BBB-on-chip systems have provided fundamental functionalities of the BBB and facilitated the testing of drug delivery to the brain tissue. However, developing biomimetic and predictive BBB models capable of reasonably mimicking essential characteristics of the BBB functions is still a challenge. In addition, detailed analysis of the dynamics of drug delivery to the healthy or diseased brain requires not only biomimetic BBB tissue models but also new systems capable of monitoring the BBB microenvironment and dynamics of barrier function and delivery mechanisms. This review provides a comprehensive overview of recent advances in microengineering of BBB models with different functional complexity and mimicking capability of healthy and diseased states. It also discusses new technologies that can make the next generation of biomimetic human BBBs containing integrated biosensors for real-time monitoring the tissue microenvironment and barrier function and correlating it with the dynamics of drug delivery. Such integrated system addresses important brain drug delivery questions related to the treatment of brain diseases. We further discuss how the combination of in vitro BBB systems, computational models and nanotechnology supports for characterization of the dynamics of drug delivery to the brain.
Collapse
Affiliation(s)
- Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - Mohsen Janmaleki
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - Mana Novin
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - John Saliba
- Biomedical Engineering, Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Fatima El-Hajj
- Biomedical Engineering, Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Mahdi RezayatiCharan
- Breast Cancer Research Center (BCRC), ACECR, Tehran, Iran; School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Amir Seyfoori
- Breast Cancer Research Center (BCRC), ACECR, Tehran, Iran; School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hamid Sadabadi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada
| | - Milène Vandal
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Minh Dang Nguyen
- Departments of Clinical Neurosciences, Cell Biology and Anatomy, Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Anwarul Hasan
- Biomedical Engineering, Department of Mechanical Engineering, Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon; Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, 2713, Qatar
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, Canada.
| |
Collapse
|
30
|
Jamieson JJ, Searson PC, Gerecht S. Engineering the human blood-brain barrier in vitro. J Biol Eng 2017; 11:37. [PMID: 29213304 PMCID: PMC5713119 DOI: 10.1186/s13036-017-0076-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
The blood-brain barrier (BBB) is the interface between the vasculature and the brain, regulating molecular and cellular transport into the brain. Endothelial cells (ECs) that form the capillary walls constitute the physical barrier but are dependent on interactions with other cell types. In vitro models are widely used in BBB research for mechanistic studies and drug screening. Current models have both biological and technical limitations. Here we review recent advances in stem cell engineering that have been utilized to create innovative platforms to replicate key features of the BBB. The development of human in vitro models is envisioned to enable new mechanistic investigations of BBB transport in central nervous system diseases.
Collapse
Affiliation(s)
- John J Jamieson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA.,Institute for Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA
| | - Peter C Searson
- Institute for Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA.,Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA.,Institute for Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA.,Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218 USA
| |
Collapse
|
31
|
Gene Expression Changes in Long-Term In Vitro Human Blood-Brain Barrier Models and Their Dependence on a Transwell Scaffold Material. JOURNAL OF HEALTHCARE ENGINEERING 2017; 2017:5740975. [PMID: 29317995 PMCID: PMC5727720 DOI: 10.1155/2017/5740975] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/19/2017] [Accepted: 10/12/2017] [Indexed: 12/24/2022]
Abstract
Disruption of the blood-brain barrier (BBB) is the hallmark of many neurovascular disorders, making it a critically important focus for therapeutic options. However, testing the effects of either drugs or pathological agents is difficult due to the potentially damaging consequences of altering the normal brain microenvironment. Recently, in vitro coculture tissue models have been developed as an alternative to animal testing. Despite low cost, these platforms use synthetic scaffolds which prevent normal barrier architecture, cellular crosstalk, and tissue remodeling. We created a biodegradable electrospun gelatin mat "biopaper" (BP) as a scaffold material for an endothelial/astrocyte coculture model allowing cell-cell contact and crosstalk. To compare the BP and traditional models, we investigated the expression of 27 genes involved in BBB permeability, cellular function, and endothelial junctions at different time points. Gene expression levels demonstrated higher expression of transcripts involved in endothelial junction formation, including TJP2 and CDH5, in the BP model. The traditional model had higher expression of genes associated with extracellular matrix-associated proteins, including SPARC and COL4A1. Overall, the results demonstrate that the BP coculture model is more representative of a healthy BBB state, though both models have advantages that may be useful in disease modeling.
Collapse
|
32
|
Maherally Z, Fillmore HL, Tan SL, Tan SF, Jassam SA, Quack FI, Hatherell KE, Pilkington GJ. Real-time acquisition of transendothelial electrical resistance in an all-human, in vitro, 3-dimensional, blood-brain barrier model exemplifies tight-junction integrity. FASEB J 2017; 32:168-182. [PMID: 28883042 PMCID: PMC5731124 DOI: 10.1096/fj.201700162r] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 08/21/2017] [Indexed: 12/02/2022]
Abstract
The blood–brain barrier (BBB) consists of endothelial cells, astrocytes, and pericytes embedded in basal lamina (BL). Most in vitro models use nonhuman, monolayer cultures for therapeutic-delivery studies, relying on transendothelial electrical resistance (TEER) measurements without other tight-junction (TJ) formation parameters. We aimed to develop reliable, reproducible, in vitro 3-dimensional (3D) models incorporating relevant human, in vivo cell types and BL proteins. The 3D BBB models were constructed with human brain endothelial cells, human astrocytes, and human brain pericytes in mono-, co-, and tricultures. TEER was measured in 3D models using a volt/ohmmeter and cellZscope. Influence of BL proteins—laminin, fibronectin, collagen type IV, agrin, and perlecan—on adhesion and TEER was assessed using an electric cell-substrate impedance–sensing system. TJ protein expression was assessed by Western blotting (WB) and immunocytochemistry (ICC). Perlecan (10 µg/ml) evoked unreportedly high, in vitro TEER values (1200 Ω) and the strongest adhesion. Coculturing endothelial cells with astrocytes yielded the greatest resistance over time. ICC and WB results correlated with resistance levels, with evidence of prominent occludin expression in cocultures. BL proteins exerted differential effects on TEER, whereas astrocytes in contact yielded higher TEER values and TJ expression.—Maherally, Z., Fillmore, H. L., Tan, S. L., Tan, S. F., Jassam, S. A., Quack, F. I., Hatherell, K. E., Pilkington, G. J. Real-time acquisition of transendothelial electrical resistance in an all-human, in vitro, 3-dimensional, blood–brain barrier model exemplifies tight-junction integrity.
Collapse
Affiliation(s)
- Zaynah Maherally
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom; and
| | - Helen L Fillmore
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom; and
| | - Sim Ling Tan
- Department of Medicine, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - Suk Fei Tan
- Department of Medicine, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - Samah A Jassam
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom; and
| | - Friederike I Quack
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom; and
| | - Kathryn E Hatherell
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom; and
| | - Geoffrey J Pilkington
- Brain Tumour Research Centre, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom; and
| |
Collapse
|
33
|
Tian Z, Zhang H, Dixon J, Traphagen N, Wyatt TA, Kharbanda K, Simet Chadwick S, Kolliputi N, Allen-Gipson DS. Cigarette Smoke Impairs A 2A Adenosine Receptor Mediated Wound Repair through Up-regulation of Duox-1 Expression. Sci Rep 2017; 7:44405. [PMID: 28337995 PMCID: PMC5364501 DOI: 10.1038/srep44405] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 01/31/2017] [Indexed: 02/03/2023] Open
Abstract
Cigarette smoke (CS) exposure and intrinsic factors such as the NADPH oxidases produce high levels of reactive oxygen species (ROS), ensuing inflammatory tissue injury. We previously demonstrated that CS-generated ROS, particularly hydrogen peroxide (H2O2), impaired adenosine stimulated wound repair. We hypothesized that CS exposure modulates expression of Dual oxidase 1 (Duox-1), a NADPH oxidases known to generate H2O2. To test this hypothesis, we used human bronchial epithelial cell line Nuli-1 and C57BL/6 mice. Cells were treated with 5% CS extract (CSE) for various periods of time, and mice were exposed to whole body CS for six weeks. Both CSE and CS treatment induced increased expression of Duox-1, and silencing of Doux-1 improved the rate of cell wound repair induced by CSE treatment. Nuli-1 cells pretreated with thapsigargin but not calcium ionophore exhibited increased Duox-1 mRNA expression. CSE treatment stimulated PKCα activation, which was effectively blocked by pretreatment with diphenylene iodonium, a NADPH oxidase inhibitor. Compared to control, lungs from CS-exposed mice showed a significant increase in PKCα activity and Duox-1 expression. Collectively, the data demonstrated that CS exposure upregulates expression of Duox-1 protein. This further leads to H2O2 production and PKCα activation, inhibiting A2AAR-stimulated wound repair.
Collapse
Affiliation(s)
- Zhi Tian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa FL, USA
| | - Hui Zhang
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jendayi Dixon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa FL, USA
| | - Nicole Traphagen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa FL, USA
| | - Todd A. Wyatt
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Environmental, Agricultural, and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
- Research Service, Omaha-Western Iowa Veterans Affairs Medical Center, Omaha, NE, USA
| | - Kusum Kharbanda
- Research Service, Omaha-Western Iowa Veterans Affairs Medical Center, Omaha, NE, USA
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Nebraska Medical Center, NE, USA
| | - Samantha Simet Chadwick
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, University of South Florida, Tampa FL, USA
| | - Diane S. Allen-Gipson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa FL, USA
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, University of South Florida, Tampa FL, USA
| |
Collapse
|
34
|
Banerjee J, Shi Y, Azevedo HS. In vitro blood–brain barrier models for drug research: state-of-the-art and new perspectives on reconstituting these models on artificial basement membrane platforms. Drug Discov Today 2016; 21:1367-1386. [DOI: 10.1016/j.drudis.2016.05.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/14/2016] [Accepted: 05/31/2016] [Indexed: 12/21/2022]
|
35
|
Qosa H, Mohamed LA, Al Rihani SB, Batarseh YS, Duong QV, Keller JN, Kaddoumi A. High-Throughput Screening for Identification of Blood-Brain Barrier Integrity Enhancers: A Drug Repurposing Opportunity to Rectify Vascular Amyloid Toxicity. J Alzheimers Dis 2016; 53:1499-516. [PMID: 27392852 PMCID: PMC4992409 DOI: 10.3233/jad-151179] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that maintains brain homeostasis and protects it from free entry of chemicals, toxins, and drugs. The barrier function of the BBB is maintained mainly by capillary endothelial cells that physically separate brain from blood. Several neurological diseases, such as Alzheimer's disease (AD), are known to disrupt BBB integrity. In this study, a high-throughput screening (HTS) was developed to identify drugs that rectify/protect BBB integrity from vascular amyloid toxicity associated with AD progression. Assessing Lucifer Yellow permeation across in-vitro BBB model composed from mouse brain endothelial cells (bEnd3) grown on 96-well plate inserts was used to screen 1280 compounds of Sigma LOPAC®1280 library for modulators of bEnd3 monolayer integrity. HTS identified 62 compounds as disruptors, and 50 compounds as enhancers of the endothelial barrier integrity. From these 50 enhancers, 7 FDA approved drugs were identified with EC50 values ranging from 0.76-4.56 μM. Of these 7 drugs, 5 were able to protect bEnd3-based BBB model integrity against amyloid toxicity. Furthermore, to test the translational potential to humans, the 7 drugs were tested for their ability to rectify the disruptive effect of Aβ in the human endothelial cell line hCMEC/D3. Only 3 (etodolac, granisetron, and beclomethasone) out of the 5 effective drugs in the bEnd3-based BBB model demonstrated a promising effect to protect the hCMEC/D3-based BBB model integrity. These drugs are compelling candidates for repurposing as therapeutic agents that could rectify dysfunctional BBB associated with AD.
Collapse
Affiliation(s)
- Hisham Qosa
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Loqman A. Mohamed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Sweilem B. Al Rihani
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Yazan S. Batarseh
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Quoc-Viet Duong
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Jeffrey N. Keller
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA, USA
| |
Collapse
|
36
|
Aman J, Weijers EM, van Nieuw Amerongen GP, Malik AB, van Hinsbergh VWM. Using cultured endothelial cells to study endothelial barrier dysfunction: Challenges and opportunities. Am J Physiol Lung Cell Mol Physiol 2016; 311:L453-66. [PMID: 27343194 DOI: 10.1152/ajplung.00393.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 06/20/2016] [Indexed: 12/24/2022] Open
Abstract
Despite considerable progress in the understanding of endothelial barrier regulation and the identification of approaches that have the potential to improve endothelial barrier function, no drug- or stem cell-based therapy is presently available to reverse the widespread vascular leak that is observed in acute respiratory distress syndrome (ARDS) and sepsis. The translational gap suggests a need to develop experimental approaches and tools that better mimic the complex environment of the microcirculation in which the vascular leak develops. Recent studies have identified several elements of this microenvironment. Among these are composition and stiffness of the extracellular matrix, fluid shear stress, interaction of endothelial cells (ECs) with pericytes, oxygen tension, and the combination of toxic and mechanic injurious stimuli. Development of novel cell culture techniques that integrate these elements would allow in-depth analysis of EC biology that closely approaches the (patho)physiological conditions in situ. In parallel, techniques to isolate organ-specific ECs, to define EC heterogeneity in its full complexity, and to culture patient-derived ECs from inducible pluripotent stem cells or endothelial progenitor cells are likely to advance the understanding of ARDS and lead to development of therapeutics. This review 1) summarizes the advantages and pitfalls of EC cultures to study vascular leak in ARDS, 2) provides an overview of elements of the microvascular environment that can directly affect endothelial barrier function, and 3) discusses alternative methods to bridge the gap between basic research and clinical application with the intent of improving the translational value of present EC culture approaches.
Collapse
Affiliation(s)
- Jurjan Aman
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands; Department of Pulmonary Diseases, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands;
| | - Ester M Weijers
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Geerten P van Nieuw Amerongen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Asrar B Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Victor W M van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Zobel K, Hansen U, Galla HJ. Blood-brain barrier properties in vitro depend on composition and assembly of endogenous extracellular matrices. Cell Tissue Res 2016; 365:233-45. [DOI: 10.1007/s00441-016-2397-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 03/14/2016] [Indexed: 01/03/2023]
|
38
|
Pandey PK, Sharma AK, Gupta U. Blood brain barrier: An overview on strategies in drug delivery, realistic in vitro modeling and in vivo live tracking. Tissue Barriers 2016; 4:e1129476. [PMID: 27141418 PMCID: PMC4836458 DOI: 10.1080/21688370.2015.1129476] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 12/25/2022] Open
Abstract
Blood brain barrier (BBB) is a group of astrocytes, neurons and endothelial cells, which makes restricted passage of various biological or chemical entities to the brain tissue. It gives protection to brain at one hand, but at the other hand it has very selective permeability for bio-actives and other foreign materials and is one of the major challenges for the drug delivery. Nanocarriers are promising to cross BBB utilizing alternative route of administration such as intranasal and intra-carotid drug delivery which bypasses BBB. In future more optimized drug delivery system can be achieved by compiling the best routes with the best carriers. Single photon emission tomography (SPECT) and different brain-on-a-chip in vitro models are being very reliable to study live in vivo tracking of BBB and its pathophysiology, respectively. In the current review we have tried to exploit mechanistically all these to understand and manage the various BBB disruptions in diseased condition along with crossing the hurdles occurring in drug or gene delivery across BBB.
Collapse
Affiliation(s)
- Pawan Kumar Pandey
- Department of Pharmacy; School of Chemical Sciences and Pharmacy, Central University of Rajasthan; Ajmer; Rajasthan, India
| | - Ashok Kumar Sharma
- Department of Pharmacy; School of Chemical Sciences and Pharmacy, Central University of Rajasthan; Ajmer; Rajasthan, India
| | - Umesh Gupta
- Department of Pharmacy; School of Chemical Sciences and Pharmacy, Central University of Rajasthan; Ajmer; Rajasthan, India
| |
Collapse
|
39
|
Serlin Y, Shelef I, Knyazer B, Friedman A. Anatomy and physiology of the blood-brain barrier. Semin Cell Dev Biol 2015; 38:2-6. [PMID: 25681530 DOI: 10.1016/j.semcdb.2015.01.002] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/07/2015] [Indexed: 02/09/2023]
Abstract
Essential requisite for the preservation of normal brain activity is to maintain a narrow and stable homeostatic control in the neuronal environment of the CNS. Blood flow alterations and altered vessel permeability are considered key determinants in the pathophysiology of brain injuries. We will review the present-day literature on the anatomy, development and physiological mechanisms of the blood-brain barrier, a distinctive and tightly regulated interface between the CNS and the peripheral circulation, playing a crucial role in the maintenance of the strict environment required for normal brain function.
Collapse
Affiliation(s)
- Yonatan Serlin
- Departments of Physiology, Cell Biology and Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ilan Shelef
- Department of Radiology, Soroka University Medical Center, Ben Gurion University of the Negev, 84101 Beer-Sheva, Israel
| | - Boris Knyazer
- Department of Ophthalmology, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Friedman
- Departments of Physiology, Cell Biology and Cognitive and Brain Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, PO Box 15000, 5850 College Street, Halifax Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
40
|
Wolff A, Antfolk M, Brodin B, Tenje M. In Vitro Blood-Brain Barrier Models-An Overview of Established Models and New Microfluidic Approaches. J Pharm Sci 2015; 104:2727-46. [PMID: 25630899 DOI: 10.1002/jps.24329] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 12/25/2022]
Abstract
The societal need for new central nervous system (CNS) medicines is substantial, because of the global increase in life expectancy and the accompanying increase in age-related CNS diseases. Low blood-brain barrier (BBB) permeability has been one of the major causes of failure for new CNS drug candidates. There has therefore been a great interest in cell models, which mimic BBB permeation properties. In this review, we present an overview of the performance of monocultured, cocultured, and triple-cultured primary cells and immortalized cell lines, including key parameters such as transendothelial electrical resistance values, permeabilities of paracellular flux markers, and expression of BBB-specific marker proteins. Microfluidic systems are gaining ground as a new automated technical platform for cell culture and systematic analysis. The performance of these systems was compared with current state-of-the-art models and it was noted that, although they show great promise, these systems have not yet reached beyond the proof-of-concept stage. In general, it was found that there were large variations in experimental protocols, BBB phenotype markers, and paracellular flux markers used. It is the author's opinion that the field may benefit greatly from developing standardized methodologies and initiating collaborative efforts on optimizing culture protocols.
Collapse
Affiliation(s)
- Anette Wolff
- Lund University, Department of Biomedical Engineering, Lund, Sweden
| | - Maria Antfolk
- Lund University, Department of Biomedical Engineering, Lund, Sweden
| | - Birger Brodin
- University of Copenhagen, Department of Pharmacy, Copenhagen, Denmark
| | - Maria Tenje
- Lund University, Department of Biomedical Engineering, Lund, Sweden.,Uppsala University, Department of Engineering Sciences, Uppsala, Sweden
| |
Collapse
|
41
|
Li J, Zhang K, Wu J, Liao Y, Yang P, Huang N. Co-culture of endothelial cells and patterned smooth muscle cells on titanium: Construction with high density of endothelial cells and low density of smooth muscle cells. Biochem Biophys Res Commun 2015; 456:555-61. [DOI: 10.1016/j.bbrc.2014.10.127] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/25/2014] [Indexed: 12/31/2022]
|
42
|
Stanimirovic DB, Bani-Yaghoub M, Perkins M, Haqqani AS. Blood-brain barrier models: in vitro to in vivo translation in preclinical development of CNS-targeting biotherapeutics. Expert Opin Drug Discov 2014; 10:141-55. [PMID: 25388782 DOI: 10.1517/17460441.2015.974545] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The majority of therapeutics, small molecule or biologics, developed for the CNS do not penetrate the blood-brain barrier (BBB) sufficiently to induce pharmacologically meaningful effects on CNS targets. To improve the efficiency of CNS drug discovery, several in vitro models of the BBB have been used to aid early selection of molecules with CNS exposure potential. However, correlative studies suggest relatively poor predictability of in vitro BBB models underscoring the need to combine in vitro and in vivo BBB penetration assessment into an integrated preclinical workflow. AREAS COVERED This review gives a brief general overview of in vitro and in vivo BBB models used in the pre-clinical evaluation of CNS-targeting drugs, with particular focus on the recent progress in developing humanized models. The authors discuss the advantages, limitations, in vitro-in vivo correlation, and integration of these models into CNS drug discovery and development with the aim of improving translation. EXPERT OPINION Often, a simplistic rationalization of the CNS drug discovery and development process overlooks or even ignores the need for an early and predictive assessment of the BBB permeability. Indeed, past failures of CNS candidates in clinical trials argue strongly that the early deployment of in vitro and in vivo models for assessing BBB permeability, mechanisms of transport and brain exposure of leads, and the co-development of BBB delivery strategies will improve translation and increase the clinical success of CNS pipelines.
Collapse
Affiliation(s)
- Danica B Stanimirovic
- Human Health Therapeutics Portfolio, National Research Council of Canada , 1200 Montreal Road, Bldg M-54 Ottawa, ON K4P 1R7 , Canada +1 613 993 3730 ; +1 613 941 4475 ;
| | | | | | | |
Collapse
|
43
|
Steiner E, Enzmann GU, Lyck R, Lin S, Rüegg MA, Kröger S, Engelhardt B. The heparan sulfate proteoglycan agrin contributes to barrier properties of mouse brain endothelial cells by stabilizing adherens junctions. Cell Tissue Res 2014; 358:465-79. [PMID: 25107608 PMCID: PMC4210653 DOI: 10.1007/s00441-014-1969-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 07/17/2014] [Indexed: 01/13/2023]
Abstract
Barrier characteristics of brain endothelial cells forming the blood–brain barrier (BBB) are tightly regulated by cellular and acellular components of the neurovascular unit. During embryogenesis, the accumulation of the heparan sulfate proteoglycan agrin in the basement membranes ensheathing brain vessels correlates with BBB maturation. In contrast, loss of agrin deposition in the vasculature of brain tumors is accompanied by the loss of endothelial junctional proteins. We therefore wondered whether agrin had a direct effect on the barrier characteristics of brain endothelial cells. Agrin increased junctional localization of vascular endothelial (VE)-cadherin, β-catenin, and zonula occludens-1 (ZO-1) but not of claudin-5 and occludin in the brain endothelioma cell line bEnd5 without affecting the expression levels of these proteins. This was accompanied by an agrin-induced reduction of the paracellular permeability of bEnd5 monolayers. In vivo, the lack of agrin also led to reduced junctional localization of VE-cadherin in brain microvascular endothelial cells. Taken together, our data support the notion that agrin contributes to barrier characteristics of brain endothelium by stabilizing the adherens junction proteins VE-cadherin and β-catenin and the junctional protein ZO-1 to brain endothelial junctions.
Collapse
Affiliation(s)
- Esther Steiner
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
44
|
Liu Q, Wu C, Cai H, Hu N, Zhou J, Wang P. Cell-based biosensors and their application in biomedicine. Chem Rev 2014; 114:6423-61. [PMID: 24905074 DOI: 10.1021/cr2003129] [Citation(s) in RCA: 189] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Qingjun Liu
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of the Ministry of Education, Department of Biomedical Engineering, Zhejiang University , Hangzhou 310027, China
| | | | | | | | | | | |
Collapse
|
45
|
Influence of basement membrane proteins and endothelial cell-derived factors on the morphology of human fetal-derived astrocytes in 2D. PLoS One 2014; 9:e92165. [PMID: 24647106 PMCID: PMC3960172 DOI: 10.1371/journal.pone.0092165] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/19/2014] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are the most prevalent type of glial cell in the brain, participating in a variety of diverse functions from regulating cerebral blood flow to controlling synapse formation. Astrocytes and astrocyte-conditioned media are widely used in models of the blood-brain barrier (BBB), however, very little is known about astrocyte culture in 2D. To test the hypothesis that surface coating and soluble factors influence astrocyte morphology in 2D, we quantitatively analyzed the morphology of human fetal derived astrocytes on glass, matrigel, fibronectin, collagen IV, and collagen I, and after the addition soluble factors including platelet-derived growth factor (PDGF), laminin, basic fibroblast growth factor (bFGF), and leukemia inhibitory factor (LIF). Matrigel surface coatings, as well as addition of leukemia inhibitory factor (LIF) to the media, were found to have the strongest effects on 2D astrocyte morphology, and may be important in improving existing BBB models. In addition, the novel set of quantitative parameters proposed in this paper provide a test for determining the influence of compounds on astrocyte morphology, both to screen for new endothelial cell-secreted factors that influence astrocytes, and to determine in a high-throughput way which factors are important for translation to more complex, 3D BBB models.
Collapse
|
46
|
Experimental tools to monitor the dynamics of endothelial barrier function: a survey of in vitro approaches. Cell Tissue Res 2014; 355:485-514. [DOI: 10.1007/s00441-014-1810-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/13/2014] [Indexed: 02/05/2023]
|
47
|
Engelhardt S, Patkar S, Ogunshola OO. Cell-specific blood-brain barrier regulation in health and disease: a focus on hypoxia. Br J Pharmacol 2014; 171:1210-30. [PMID: 24641185 PMCID: PMC3952799 DOI: 10.1111/bph.12489] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/02/2013] [Accepted: 10/16/2013] [Indexed: 01/16/2023] Open
Abstract
The blood-brain barrier (BBB) is a complex vascular structure consisting of microvascular endothelial cells that line the vessel wall, astrocyte end-feet, pericytes, as well as the basal lamina. BBB cells act in concert to maintain the characteristic impermeable and low paracellular flux of the brain vascular network, thus ensuring a homeostatic neuronal environment. Alterations in BBB stability that occur during injury have dire consequences on disease progression and it is clear that BBB cell-specific responses, positive or negative, must make a significant contribution to injury outcome. Reduced oxygenation, or hypoxia, is a characteristic of many brain diseases that significantly increases barrier permeability. Recent data suggest that hypoxia-inducible factor (HIF-1), the master regulator of the hypoxic response, probably mediates many hypoxic effects either directly or indirectly via its target genes. This review discusses current knowledge of physiological cell-specific regulation of barrier function, their responses to hypoxia as well as consequences of hypoxic- and HIF-1-mediated mechanisms on barrier integrity during select brain diseases. In the final sections, the potential of current advances in targeting HIF-1 as a therapeutic strategy will be overviewed.
Collapse
Affiliation(s)
- S Engelhardt
- Institute of Veterinary Physiology, University of ZurichZurich, Switzerland
| | - S Patkar
- Institute of Veterinary Physiology, University of ZurichZurich, Switzerland
| | - O O Ogunshola
- Institute of Veterinary Physiology, University of ZurichZurich, Switzerland
| |
Collapse
|
48
|
Premaraj T, Simet S, Beatty M, Premaraj S. Oral epithelial cell reaction after exposure to Invisalign plastic material. Am J Orthod Dentofacial Orthop 2014; 145:64-71. [PMID: 24373656 DOI: 10.1016/j.ajodo.2013.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/01/2013] [Accepted: 09/01/2013] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Invisalign plastic aligners (Align Technology, Santa Clara, Calif) are used to correct malocclusions. The aligners wrap around the teeth and are in contact with gingival epithelium during treatment. The purpose of this study was to evaluate the cellular responses of oral epithelium exposed to Invisalign plastic in vitro. METHODS Oral epithelial cells were exposed to eluate obtained by soaking Invisalign plastic in either saline solution or artificial saliva for 2, 4, and 8 weeks. Cells grown in media containing saline solution or saliva served as controls. Morphologic changes were assessed by light microscopy. The 3-[4, 5-dimethythiazol- 2-yl]-2, 5-diphenyl tetrazolium bromide assay and flow cytometry were used to determine cell viability and membrane integrity, respectively. Cellular adhesion and micromotion of epithelial cells were measured in real time by electrical cell-substrate impedance sensing. RESULTS Cells exposed to saline-solution eluate appeared rounded, were lifted from the culture plates, and demonstrated significantly increased metabolic inactivity or cell death (P <0.05). Saliva eluates did not induce significant changes in cell viability compared with untreated cells. Flow cytometry and electric cell-substrate impedance sensing showed that cells treated with saline-solution eluate exhibited compromised membrane integrity, and reduced cell-to-cell contact and mobility when compared with saliva-eluate treatment. CONCLUSIONS Exposure to Invisalign plastic caused changes in viability, membrane permeability, and adhesion of epithelial cells in a saline-solution environment. Microleakage and hapten formation secondary to compromised epithelial integrity might lead to isocyanate allergy, which could be systemic or localized to gingiva. However, these results suggest that saliva might offer protection.
Collapse
Affiliation(s)
- Thyagaseely Premaraj
- Assistant professor, Orthodontic Section, Department of Growth and Development, College of Dentistry, University of Nebraska Medical Center, Lincoln, Neb.
| | - Samantha Simet
- Postdoctoral research associate, Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center, Omaha, Neb
| | - Mark Beatty
- Associate professor and graduate program director, Orthodontic Section, Department of Growth and Development, College of Dentistry, University of Nebraska Medical Center, Lincoln, Neb
| | - Sundaralingam Premaraj
- Professor and director, Section of Biomaterials, Department of Adult Restorative Dentistry, College of Dentistry, University of Nebraska Medical Center, Lincoln, Neb
| |
Collapse
|
49
|
Palmer AM, Alavijeh MS. Overview of experimental models of the blood-brain barrier in CNS drug discovery. ACTA ACUST UNITED AC 2013; 62:7.15.1-7.15.30. [PMID: 24510719 DOI: 10.1002/0471141755.ph0715s62] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The blood-brain barrier (BBB) is a physical and metabolic entity that isolates the brain from the systemic circulation. The barrier consists of tight junctions between endothelial cells that contain egress transporters and catabolic enzymes. To cross the BBB, a drug must possess the appropriate physicochemical properties to achieve a sufficient time-concentration profile in brain interstitial fluid (ISF). In this overview, we review techniques to measure BBB permeation, which is evidenced by the free concentration of compound in brain ISF over time. We consider a number of measurement techniques, including in vivo microdialysis and brain receptor occupancy following perfusion. Consideration is also given to the endothelial and nonendothelial cell systems used to assess both the BBB permeation of a test compound and its interactions with egress transporters, and computer models employed for predicting passive permeation and the probability of interactions with BBB transporters.
Collapse
|
50
|
Schenk GJ, Dijkstra S, van het Hof AJ, van der Pol SMA, Drexhage JAR, van der Valk P, Reijerkerk A, van Horssen J, de Vries HE. Roles for HB-EGF and CD9 in multiple sclerosis. Glia 2013; 61:1890-905. [PMID: 24038577 DOI: 10.1002/glia.22565] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 07/24/2013] [Accepted: 07/29/2013] [Indexed: 11/06/2022]
Abstract
Early events in multiple sclerosis (MS) lesion formation are loss of blood-brain barrier (BBB) integrity, immune cell trafficking into the central nervous system, and demyelination. To date, the molecular mechanisms underlying these pathogenic events are poorly understood. Heparin-binding epidermal growth factor (HB-EGF) is a trophic factor that is induced by inflammatory stimuli and has previously been shown to interact with tetraspanins (TSPs), a family of transmembrane proteins that are involved in cellular migration and adhesion. Given the known roles of TSPs and HB-EGF, we hypothesized that HB-EGF and TSPs may play a role in the processes that underlie MS lesion formation. We examined the expression of HB-EGF and the TSPs CD9 and CD81 in MS brain and found that HB-EGF was highly induced in reactive astrocytes in active lesions. TSPs were constitutively expressed throughout normal appearing white matter and control white matter. In contrast, CD9 was reduced in demyelinated lesions and increased on blood vessels in lesion areas. In vitro studies revealed that expression of HB-EGF and TSPs is regulated during inflammation. Importantly, blocking either HB-EGF or CD9 significantly reduced the migration of monocytes across brain endothelial cell monolayers. Moreover, blocking CD9 strongly enhanced the barrier function of the BBB in vitro. Together, we demonstrate that these molecules are likely implicated in processes that are highly relevant for MS lesion formation, and therefore, HB-EGF and TSPs are promising therapeutic targets.
Collapse
Affiliation(s)
- Geert J Schenk
- Department of Anatomy and Neurosciences, Section of Clinical Neuroscience, VU University Medical Center, Amsterdam, The Netherlands; Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|