1
|
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
|
2
|
Zhang L, Zhu B, Zhou X, Ning H, Zhang F, Yan B, Chen J, Ma T. ZNF787 and HDAC1 Mediate Blood-Brain Barrier Permeability in an In Vitro Model of Alzheimer's Disease Microenvironment. Neurotox Res 2024; 42:12. [PMID: 38329647 DOI: 10.1007/s12640-024-00693-4] [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: 05/31/2023] [Revised: 12/19/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
The permeability of the blood-brain barrier (BBB) is increased in Alzheimer's disease (AD). This plays a key role in the instigation and maintenance of chronic inflammation during AD. Experiments using AD models showed that the increased permeability of the BBB was mainly caused by the decreased expression of tight junction-related proteins occludin and claudin-5. In this study, we found that ZNF787 and HDAC1 were upregulated in β-amyloid (Aβ)1-42-incubated endothelial cells, resulting in increased BBB permeability. Conversely, the silencing of ZNF787 and HDAC1 by RNAi led to reduced BBB permeability. The silencing of ZNF787 and HDAC1 enhanced the expression of occludin and claudin-5. Mechanistically, ZNF787 binds to promoter regions for occludin and claudin-5 and functions as a transcriptional regulator. Furthermore, we demonstrate that ZNF787 interacts with HDAC1, and this resulted in the downregulation of the expression of genes encoding tight junction-related proteins to increase in BBB permeability. Taken together, our study identifies critical roles for the interaction between ZNF787 and HDAC1 in regulating BBB permeability and the pathogenesis of AD.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Baicheng Zhu
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Xinxin Zhou
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110034, China
| | - Hao Ning
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Fengying Zhang
- Department of Neurology, Affiliated Nanhua Hospital, University of South China, Hengyang, 421001, China
| | - Bingju Yan
- Department of Cardiology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Jiajia Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Teng Ma
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
3
|
Ozgür B, Puris E, Brachner A, Appelt-Menzel A, Oerter S, Balzer V, Holst MR, Christiansen RF, Hyldig K, Buckley ST, Kristensen M, Auriola S, Jensen A, Fricker G, Nielsen MS, Neuhaus W, Brodin B. Characterization of an iPSC-based barrier model for blood-brain barrier investigations using the SBAD0201 stem cell line. Fluids Barriers CNS 2023; 20:96. [PMID: 38115090 PMCID: PMC10731806 DOI: 10.1186/s12987-023-00501-9] [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: 10/17/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) models based on primary murine, bovine, and porcine brain capillary endothelial cell cultures have long been regarded as robust models with appropriate properties to examine the functional transport of small molecules. However, species differences sometimes complicate translating results from these models to human settings. During the last decade, brain capillary endothelial-like cells (BCECs) have been generated from stem cell sources to model the human BBB in vitro. The aim of the present study was to establish and characterize a human BBB model using human induced pluripotent stem cell (hiPSC)-derived BCECs from the hIPSC line SBAD0201. METHODS The model was evaluated using transcriptomics, proteomics, immunocytochemistry, transendothelial electrical resistance (TEER) measurements, and, finally, transport assays to assess the functionality of selected transporters and receptor (GLUT-1, LAT-1, P-gp and LRP-1). RESULTS The resulting BBB model displayed an average TEER of 5474 ± 167 Ω·cm2 and cell monolayer formation with claudin-5, ZO-1, and occludin expression in the tight junction zones. The cell monolayers expressed the typical BBB markers VE-cadherin, VWF, and PECAM-1. Transcriptomics and quantitative targeted absolute proteomics analyses revealed that solute carrier (SLC) transporters were found in high abundance, while the expression of efflux transporters was relatively low. Transport assays using GLUT-1, LAT-1, and LRP-1 substrates and inhibitors confirmed the functional activities of these transporters and receptors in the model. A transport assay suggested that P-gp was not functionally expressed in the model, albeit antibody staining revealed that P-gp was localized at the luminal membrane. CONCLUSIONS In conclusion, the novel SBAD0201-derived BBB model formed tight monolayers and was proven useful for studies investigating GLUT-1, LAT-1, and LRP-1 mediated transport across the BBB. However, the model did not express functional P-gp and thus is not suitable for the performance of drug efflux P-gp reletated studies.
Collapse
Affiliation(s)
- Burak Ozgür
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, DK-2500, Denmark
| | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Andreas Brachner
- AIT - Austrian Institute of Technology GmbH, Vienna, 1210, Austria
| | - Antje Appelt-Menzel
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT) Röntgenring 11, 97070, Würzburg, Germany
| | - Sabrina Oerter
- Chair Tissue Engineering and Regenerative Medicine (TERM), University Hospital Würzburg, 97070, Würzburg, Germany
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies (TLC-RT) Röntgenring 11, 97070, Würzburg, Germany
| | - Viktor Balzer
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | | | | | - Kathrine Hyldig
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, DK-2500, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, DK-8000, Denmark
| | - Stephen T Buckley
- Global Research Technologies, Novo Nordisk A/S, Måløv, DK-2760, Denmark
| | - Mie Kristensen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Allan Jensen
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, DK-2500, Denmark
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Heidelberg, Germany
| | | | - Winfried Neuhaus
- AIT - Austrian Institute of Technology GmbH, Vienna, 1210, Austria
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, Krems, 3500, Austria
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, DK-2100, Denmark.
| |
Collapse
|
4
|
Lyck R, Nishihara H, Aydin S, Soldati S, Engelhardt B. Modeling Brain Vasculature Immune Interactions In Vitro. Cold Spring Harb Perspect Med 2023; 13:a041185. [PMID: 36617644 PMCID: PMC10513158 DOI: 10.1101/cshperspect.a041185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The endothelial blood-brain barrier (BBB) protects central nervous system (CNS) neurons from the changeable milieu of the bloodstream by strictly controlling the movement of molecules and immune cells between the blood and the CNS. Immune cell migration across the vascular wall is a multistep process regulated by the sequential interaction of different signaling and adhesion molecules on the endothelium and the immune cells. Accounting for its unique barrier properties and trafficking molecule expression profile, particular adaptions in immune cell migration across the BBB have been observed. Thus, in vitro models of the BBB are desirable to explore the precise cellular and molecular mechanisms involved in immune cell trafficking across the BBB. The challenge to overcome is that barrier properties of brain microvascular endothelial cells are not intrinsic and readily lost in culture. With a focus on human in vitro BBB models, we here discuss the suitability of available in vitro models for the BBB for exploring the specific mechanisms involved in immune cell trafficking across the BBB.
Collapse
Affiliation(s)
- Ruth Lyck
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Sidar Aydin
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, CH 3012 Bern, Switzerland
| |
Collapse
|
5
|
Girard SD, Julien-Gau I, Molino Y, Combes BF, Greetham L, Khrestchatisky M, Nivet E. High and low permeability of human pluripotent stem cell-derived blood-brain barrier models depend on epithelial or endothelial features. FASEB J 2023; 37:e22770. [PMID: 36688807 DOI: 10.1096/fj.202201422r] [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: 08/31/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023]
Abstract
The search for reliable human blood-brain barrier (BBB) models represents a challenge for the development/testing of strategies aiming to enhance brain delivery of drugs. Human-induced pluripotent stem cells (hiPSCs) have raised hopes in the development of predictive BBB models. Differentiating strategies are thus required to generate endothelial cells (ECs), a major component of the BBB. Several hiPSC-based protocols have reported the generation of in vitro models with significant differences in barrier properties. We studied in depth the properties of iPSCs byproducts from two protocols that have been established to yield these in vitro barrier models. Our analysis/study reveals that iPSCs derivatives endowed with EC features yield high permeability models while the cells that exhibit outstanding barrier properties show principally epithelial cell-like (EpC) features. We found that models containing EpC-like cells express tight junction proteins, transporters/efflux pumps and display a high functional tightness with very low permeability, which are features commonly shared between BBB and epithelial barriers. Our study demonstrates that hiPSC-based BBB models need extensive characterization beforehand and that a reliable human BBB model containing EC-like cells and displaying low permeability is still needed.
Collapse
Affiliation(s)
- Stéphane D Girard
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
- Faculty of Medicine, VECT-HORUS SAS, Marseille, France
| | | | - Yves Molino
- Faculty of Medicine, VECT-HORUS SAS, Marseille, France
| | | | - Louise Greetham
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
| | - Michel Khrestchatisky
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
| | - Emmanuel Nivet
- Institute of NeuroPhysiopathology, INP, CNRS, Aix-Marseille University, Marseille, France
| |
Collapse
|
6
|
Quantitative Targeted Absolute Proteomics for Better Characterization of an In Vitro Human Blood-Brain Barrier Model Derived from Hematopoietic Stem Cells. Cells 2022; 11:cells11243963. [PMID: 36552728 PMCID: PMC9776576 DOI: 10.3390/cells11243963] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
We previously developed an in vitro model of the human blood-brain barrier (BBB) based on the use of endothelial cells derived from CD34+-hematopoietic stem cells and cultured with brain pericytes. The purpose of the present study was to provide information on the protein expression levels of the transporters, receptors, tight junction/adherence junction molecules, and transporter-associated molecules of human brain-like endothelial cells (hBLECs). The absolute protein expression levels were determined by liquid chromatography-mass spectrometry-based quantitative targeted absolute proteomics and compared with those from human brain microvessels (hBMVs). The protein levels of CD144, CD147, MRP4, Annexin A6 and caveolin-1 showed more than 3-fold abundance in hBLECs, those of MCT1, Connexin 43, TfR1, and claudin-5 showed less than 3-fold differences, and the protein levels of other drug efflux transporters and nutrient transporters were less represented in hBLECs than in hBMVs. It is noteworthy that BCRP was more expressed than MDR1 in hBLECs, as this was the case for hBMVs. These results suggest that transports mediated by MCT1, TfR1, and claudin-5-related tight junction function reflect the in vivo BBB situation. The present study provided a better characterization of hBLECs and clarified the equivalence of the transport characteristics between in vitro BBB models and in vivo BBB models using LC-MS/MS-based protein quantification.
Collapse
|
7
|
Smith BC, Tinkey RA, Shaw BC, Williams JL. Targetability of the neurovascular unit in inflammatory diseases of the central nervous system. Immunol Rev 2022; 311:39-49. [PMID: 35909222 PMCID: PMC9489669 DOI: 10.1111/imr.13121] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) is a selectively permeable barrier separating the periphery from the central nervous system (CNS). The BBB restricts the flow of most material into and out of the CNS, including many drugs that could be used as potent therapies. BBB permeability is modulated by several cells that are collectively called the neurovascular unit (NVU). The NVU consists of specialized CNS endothelial cells (ECs), pericytes, astrocytes, microglia, and neurons. CNS ECs maintain a complex "seal" via tight junctions, forming the BBB; breakdown of these tight junctions leads to BBB disruption. Pericytes control the vascular flow within capillaries and help maintain the basal lamina. Astrocytes control much of the flow of material that has moved beyond the CNS EC layer and can form a secondary barrier under inflammatory conditions. Microglia survey the border of the NVU for noxious material. Neuronal activity also plays a role in the maintenance of the BBB. Since astrocytes, pericytes, microglia, and neurons are all able to modulate the permeability of the BBB, understating the complex contributions of each member of the NVU will potentially uncover novel and effective methods for delivery of neurotherapies to the CNS.
Collapse
Affiliation(s)
- Brandon C. Smith
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Department of Biological, Geological, and Environmental SciencesCleveland State UniversityClevelandOhioUSA
| | - Rachel A. Tinkey
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,School of Biomedical SciencesKent State UniversityKentOhioUSA
| | - Benjamin C. Shaw
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Jessica L. Williams
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Brain Health Research Institute, Kent State UniversityKentOhioUSA
| |
Collapse
|
8
|
He T, Yang GY, Zhang Z. Crosstalk of Astrocytes and Other Cells during Ischemic Stroke. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060910. [PMID: 35743941 PMCID: PMC9228674 DOI: 10.3390/life12060910] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Astrocytes structurally compose tripartite synapses, blood–brain barrier, and the neurovascular unit and perform multiple functions through cell-to-cell signaling of neurons, glial cells, and vasculature. The crosstalk of astrocytes and other cells is complicated and incompletely understood. Here we review the role of astrocytes in response to ischemic stroke, both beneficial and detrimental, from a cell–cell interaction perspective. Reactive astrocytes provide neuroprotection through antioxidation and antiexcitatory effects and metabolic support; they also contribute to neurorestoration involving neurogenesis, synaptogenesis, angiogenesis, and oligodendrogenesis by crosstalk with stem cells and cell lineage. In the meantime, reactive astrocytes also play a vital role in neuroinflammation and brain edema. Glial scar formation in the chronic phase hinders functional recovery. We further discuss astrocyte enriched microRNAs and exosomes in the regulation of ischemic stroke. In addition, the latest notion of reactive astrocyte subsets and astrocytic activity revealed by optogenetics is mentioned. This review discusses the current understanding of the intimate molecular conversation between astrocytes and other cells and outlines its potential implications after ischemic stroke. “Neurocentric” strategies may not be sufficient for neurological protection and recovery; future therapeutic strategies could target reactive astrocytes.
Collapse
Affiliation(s)
- Tingting He
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China;
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| |
Collapse
|
9
|
Cheng Y, Medina A, Yao Z, Basu M, Natekar JP, Lang J, Sanchez E, Nkembo MB, Xu C, Qian X, Nguyen PTT, Wen Z, Song H, Ming GL, Kumar M, Brinton MA, Li MMH, Tang H. Intrinsic antiviral immunity of barrier cells revealed by an iPSC-derived blood-brain barrier cellular model. Cell Rep 2022; 39:110885. [PMID: 35649379 PMCID: PMC9230077 DOI: 10.1016/j.celrep.2022.110885] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/27/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
Physiological blood-tissue barriers play a critical role in separating the circulation from immune-privileged sites and denying access to blood-borne viruses. The mechanism of virus restriction by these barriers is poorly understood. We utilize induced pluripotent stem cell (iPSC)-derived human brain microvascular endothelial cells (iBMECs) to study virus-blood-brain barrier (BBB) interactions. These iPSC-derived cells faithfully recapitulate a striking difference in in vivo neuroinvasion by two alphavirus isolates and are selectively permissive to neurotropic flaviviruses. A model of cocultured iBMECs and astrocytes exhibits high transendothelial electrical resistance and blocks non-neurotropic flaviviruses from getting across the barrier. We find that iBMECs constitutively express an interferon-induced gene, IFITM1, which preferentially restricts the replication of non-neurotropic flaviviruses. Barrier cells from blood-testis and blood-retinal barriers also constitutively express IFITMs that contribute to the viral resistance. Our application of a renewable human iPSC-based model for studying virus-BBB interactions reveals that intrinsic immunity at the barriers contributes to virus exclusion. Using a stem cell-derived cellular model and a panel of human pathogenic viruses, Cheng et al. show a mechanism by which some viruses can penetrate the blood-brain barrier and cause diseases in the central nervous system.
Collapse
Affiliation(s)
- Yichen Cheng
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Angelica Medina
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Zhenlan Yao
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Mausumi Basu
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Jianshe Lang
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Egan Sanchez
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Mezindia B Nkembo
- Department of Biological Science, Florida State University, Tallahassee, FL, USA
| | - Chongchong Xu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Xuyu Qian
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Phuong T T Nguyen
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Hongjun Song
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mukesh Kumar
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Margo A Brinton
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Melody M H Li
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
10
|
Wan J, Zhou S, Mea HJ, Guo Y, Ku H, Urbina BM. Emerging Roles of Microfluidics in Brain Research: From Cerebral Fluids Manipulation to Brain-on-a-Chip and Neuroelectronic Devices Engineering. Chem Rev 2022; 122:7142-7181. [PMID: 35080375 DOI: 10.1021/acs.chemrev.1c00480] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Remarkable progress made in the past few decades in brain research enables the manipulation of neuronal activity in single neurons and neural circuits and thus allows the decipherment of relations between nervous systems and behavior. The discovery of glymphatic and lymphatic systems in the brain and the recently unveiled tight relations between the gastrointestinal (GI) tract and the central nervous system (CNS) further revolutionize our understanding of brain structures and functions. Fundamental questions about how neurons conduct two-way communications with the gut to establish the gut-brain axis (GBA) and interact with essential brain components such as glial cells and blood vessels to regulate cerebral blood flow (CBF) and cerebrospinal fluid (CSF) in health and disease, however, remain. Microfluidics with unparalleled advantages in the control of fluids at microscale has emerged recently as an effective approach to address these critical questions in brain research. The dynamics of cerebral fluids (i.e., blood and CSF) and novel in vitro brain-on-a-chip models and microfluidic-integrated multifunctional neuroelectronic devices, for example, have been investigated. This review starts with a critical discussion of the current understanding of several key topics in brain research such as neurovascular coupling (NVC), glymphatic pathway, and GBA and then interrogates a wide range of microfluidic-based approaches that have been developed or can be improved to advance our fundamental understanding of brain functions. Last, emerging technologies for structuring microfluidic devices and their implications and future directions in brain research are discussed.
Collapse
Affiliation(s)
- Jiandi Wan
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Sitong Zhou
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Hing Jii Mea
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Yaojun Guo
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Hansol Ku
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Brianna M Urbina
- Biochemistry, Molecular, Cellular and Developmental Biology Program, University of California, Davis, California 95616, United States
| |
Collapse
|
11
|
In vitro blood brain barrier models: An overview. J Control Release 2022; 343:13-30. [PMID: 35026351 DOI: 10.1016/j.jconrel.2022.01.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/22/2022]
Abstract
Understanding the composition and function of the blood brain barrier (BBB) enables the development of novel, innovative techniques for administering central nervous system (CNS) medications and technologies for improving the existing models. Scientific and methodological interest in the pathology of the BBB resulted in the formation of numerous in vitro BBB models. Once successfully studied and modelled, it would be a valuable tool for elucidating the mechanism of action of the CNS disorders prior to their manifestation and the pathogenic factors. Understanding the rationale behind the selection of the models as well as their working may enable the development of state-of-the-art drugs for treating and managing neurological diseases. Hence, to have realistic simulation of the BBB and test its drug permeability the microfluidics-based BBB-on-Chip model has been developed. To summarise, we aim to evaluate the advanced, newly developed and frequently used in vitro BBB models, thereby providing a brief overview of the components essential for in vitro BBB formation, the methods of chip fabrication and cell culturing, its applications and the recent advances in this technological field. This will be critical for developing CNS treatments with improved BBB penetrability and pharmacokinetic properties.
Collapse
|
12
|
Lachaux J, Hwang G, Arouche N, Naserian S, Harouri A, Lotito V, Casari C, Lok T, Menager JB, Issard J, Guihaire J, Denis CV, Lenting PJ, Barakat AI, Uzan G, Mercier O, Haghiri-Gosnet AM. A compact integrated microfluidic oxygenator with high gas exchange efficiency and compatibility for long-lasting endothelialization. LAB ON A CHIP 2021; 21:4791-4804. [PMID: 34309615 DOI: 10.1039/d1lc00356a] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
We have developed and tested a novel microfluidic device for blood oxygenation, which exhibits a large surface area of gas exchange and can support long-term sustainable endothelialization of blood microcapillaries, enhancing its hemocompatibility for clinical applications. The architecture of the parallel stacking of the trilayers is based on a central injection for blood and a lateral injection/output for gas which allows significant reduction in shear stress, promoting sustainable endothelialization since cells can be maintained viable for up to 2 weeks after initial seeding in the blood microchannel network. The circular design of curved blood capillaries allows covering a maximal surface area at 4 inch wafer scale, producing high oxygen uptake and carbon dioxide release in each single unit. Since the conventional bonding process based on oxygen plasma cannot be used for surface areas larger than several cm2, a new "wet bonding" process based on soft microprinting has been developed and patented. Using this new protocol, each 4 inch trilayer unit can be sealed without a collapsed membrane even at reduced 15 μm thickness and can support a high blood flow rate. The height of the blood channels has been optimized to reduce pressure drop and enhance gas exchange at a high volumetric blood flow rate up to 15 ml min-1. The simplicity of connecting different units in the stacked architecture is demonstrated for 3- or 5-unit stacked devices that exhibit remarkable performance with low primary volume, high oxygen uptake and carbon dioxide release and high flow rate of up to 80 ml min-1.
Collapse
Affiliation(s)
- Julie Lachaux
- Université Paris-Saclay, CNRS, Centre de Nanosciences et Nanotechnologies C2N, UMR9001, Palaiseau 91120, France.
| | - Gilgueng Hwang
- Université Paris-Saclay, CNRS, Centre de Nanosciences et Nanotechnologies C2N, UMR9001, Palaiseau 91120, France.
| | - Nassim Arouche
- Université Paris-Saclay, INSERM, UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
| | - Sina Naserian
- Université Paris-Saclay, INSERM, UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
| | - Abdelmounaim Harouri
- Université Paris-Saclay, CNRS, Centre de Nanosciences et Nanotechnologies C2N, UMR9001, Palaiseau 91120, France.
| | - Valeria Lotito
- Université Paris-Saclay, CNRS, Centre de Nanosciences et Nanotechnologies C2N, UMR9001, Palaiseau 91120, France.
| | - Caterina Casari
- Université Paris-Saclay, INSERM, UMR S1176, Le Kremin-Bicêtre, France
| | - Thevy Lok
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau 91120, France
| | - Jean Baptiste Menager
- Université Paris-Saclay, INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Justin Issard
- Université Paris-Saclay, INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Julien Guihaire
- Université Paris-Saclay, INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Cécile V Denis
- Université Paris-Saclay, INSERM, UMR S1176, Le Kremin-Bicêtre, France
| | - Peter J Lenting
- Université Paris-Saclay, INSERM, UMR S1176, Le Kremin-Bicêtre, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, Palaiseau 91120, France
| | - Georges Uzan
- Université Paris-Saclay, INSERM, UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
| | - Olaf Mercier
- Université Paris-Saclay, INSERM UMR_S 999 "Pulmonary Hypertension: Pathophysiology and Novel Therapies", Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Anne-Marie Haghiri-Gosnet
- Université Paris-Saclay, CNRS, Centre de Nanosciences et Nanotechnologies C2N, UMR9001, Palaiseau 91120, France.
| |
Collapse
|
13
|
Gastfriend BD, Nishihara H, Canfield SG, Foreman KL, Engelhardt B, Palecek SP, Shusta EV. Wnt signaling mediates acquisition of blood-brain barrier properties in naïve endothelium derived from human pluripotent stem cells. eLife 2021; 10:70992. [PMID: 34755601 PMCID: PMC8664294 DOI: 10.7554/elife.70992] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Endothelial cells (ECs) in the central nervous system (CNS) acquire their specialized blood-brain barrier (BBB) properties in response to extrinsic signals, with Wnt/β-catenin signaling coordinating multiple aspects of this process. Our knowledge of CNS EC development has been advanced largely by animal models, and human pluripotent stem cells (hPSCs) offer the opportunity to examine BBB development in an in vitro human system. Here we show that activation of Wnt signaling in hPSC-derived naïve endothelial progenitors, but not in matured ECs, leads to robust acquisition of canonical BBB phenotypes including expression of GLUT-1, increased claudin-5, decreased PLVAP and decreased permeability. RNA-seq revealed a transcriptome profile resembling ECs with CNS-like characteristics, including Wnt-upregulated expression of LEF1, APCDD1, and ZIC3. Together, our work defines effects of Wnt activation in naïve ECs and establishes an improved hPSC-based model for interrogation of CNS barriergenesis.
Collapse
Affiliation(s)
- Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | | | - Scott G Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | - Koji L Foreman
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | | | - Sean P Palecek
- Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| | - Eric V Shusta
- Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, United States
| |
Collapse
|
14
|
Blanco-Elices C, Chato-Astrain J, Oyonarte S, Bermejo-Casares F, España-López A, Fernández-Valadés R, Sánchez-Quevedo MDC, Alaminos M, Martín-Piedra MA, Garzón I. Generation of a novel model of bioengineered human oral mucosa with increased vascularization potential. J Periodontal Res 2021; 56:1116-1131. [PMID: 34510438 PMCID: PMC9293188 DOI: 10.1111/jre.12927] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/29/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
Objective The aim of this study was to generate novel models of bioartificial human oral mucosa with increased vascularization potential for future use as an advanced therapies medicinal product, by using different vascular and mesenchymal stem cell sources. Background Oral mucosa substitutes could contribute to the clinical treatment of complex diseases affecting the oral cavity. Although several models of artificial oral mucosa have been described, biointegration is a major issue that could be favored by the generation of novel substitutes with increased vascularization potential once grafted in vivo. Methods Three types of mesenchymal stem cells (MSCs) were obtained from adipose tissue, bone marrow, and dental pulp, and their in vitro potential was evaluated by inducing differentiation to the endothelial lineage using conditioning media. Then, 3D models of human artificial oral mucosa were generated using biocompatible fibrin‐agarose biomaterials combined with human oral mucosa fibroblasts and each type of MSC before and after induction to the endothelial lineage, using human umbilical vein endothelial cells (HUVEC) as controls. The vascularization potential of each oral mucosa substitute was assessed in vitro and in vivo in nude mice. Results In vitro induction of MSCs kept in culture was able to increase the expression of VEGF, CD31, and vWF endothelial markers, especially in bone marrow and dental pulp‐MSCs, and numerous proteins with a role in vasculogenesis become overexpressed. Then, in vivo grafting resulted in a significant increase in blood vessels formation at the interface area between the graft and the host tissues, with significantly positive expression of VEGF, CD31, vWF, and CD34 as compared to negative controls, especially when pre‐differentiated MSCs derived from bone marrow and dental pulp were used. In addition, a significantly higher number of cells committed to the endothelial lineage expressing the same endothelial markers were found within the bioartificial tissue. Conclusion Our results suggest that the use of pre‐differentiated MSCs could contribute to a rapid generation of a vascular network that may favor in vivo biointegration of bioengineered human oral mucosa substitutes.
Collapse
Affiliation(s)
- Cristina Blanco-Elices
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Doctoral Programme in Biomedicine, University of Granada, Granada, Spain
| | - Jesús Chato-Astrain
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Salvador Oyonarte
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Andalusian Network for Transfusional Medicine, Cells and Tissues and Blood and Tissue Bank of Granada, Granada, Spain
| | | | - Antonio España-López
- Craniofacial Malformations and Cleft Lip and Palate Management Unit, University Hospital Virgen de las Nieves, Granada, Spain
| | - Ricardo Fernández-Valadés
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Division of Pediatric Surgery, University Hospital Virgen de las Nieves, Granada, Spain
| | - Maria Del Carmen Sánchez-Quevedo
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miguel Alaminos
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miguel Angel Martín-Piedra
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Ingrid Garzón
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| |
Collapse
|
15
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
16
|
Nouri Barkestani M, Shamdani S, Afshar Bakshloo M, Arouche N, Bambai B, Uzan G, Naserian S. TNFα priming through its interaction with TNFR2 enhances endothelial progenitor cell immunosuppressive effect: new hope for their widespread clinical application. Cell Commun Signal 2021; 19:1. [PMID: 33397378 PMCID: PMC7784277 DOI: 10.1186/s12964-020-00683-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022] Open
Abstract
Background Bone marrow derived endothelial progenitor cells (EPCs) are immature endothelial cells (ECs) involved in neo-angiogenesis and endothelial homeostasis and are considered as a circulating reservoir for endothelial repair. Many studies showed that EPCs from patients with cardiovascular pathologies are impaired and insufficient; hence, allogenic sources of EPCs from adult or cord blood are considered as good choices for cell therapy applications. However, allogenic condition increases the chance of immune rejection, especially by T cells, before exerting the desired regenerative functions. TNFα is one of the main mediators of EPC activation that recognizes two distinct receptors, TNFR1 and TNFR2. We have recently reported that human EPCs are immunosuppressive and this effect was TNFα-TNFR2 dependent. Here, we aimed to investigate if an adequate TNFα pre-conditioning could increase TNFR2 expression and prime EPCs towards more immunoregulatory functions. Methods EPCs were pre-treated with several doses of TNFα to find the proper dose to up-regulate TNFR2 while keeping the TNFR1 expression stable. Then, co-cultures of human EPCs and human T cells were performed to assess whether TNFα priming would increase EPC immunosuppressive and immunomodulatory effect. Results Treating EPCs with 1 ng/ml TNFα significantly up-regulated TNFR2 expression without unrestrained increase of TNFR1 and other endothelial injury markers. Moreover, TNFα priming through its interaction with TNFR2 remarkably enhanced EPC immunosuppressive and anti-inflammatory effects. Conversely, blocking TNFR2 using anti-TNFR2 mAb followed by 1 ng/ml of TNFα treatment led to the TNFα-TNFR1 interaction and polarized EPCs towards pro-inflammatory and immunogenic functions. Conclusions We report for the first time the crucial impact of inflammation notably the TNFα-TNFR signaling pathway on EPC immunological function. Our work unveils the pro-inflammatory role of the TNFα-TNFR1 axis and, inversely the anti-inflammatory implication of the TNFα-TNFR2 axis in EPC immunoregulatory functions. Priming EPCs with 1 ng/ml of TNFα prior to their administration could boost them toward a more immunosuppressive phenotype. This could potentially lead to EPCs’ longer presence in vivo after their allogenic administration resulting in their better contribution to angiogenesis and vascular regeneration. Video Abstract
Collapse
Affiliation(s)
- Mahsa Nouri Barkestani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Sara Shamdani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Paris-Saclay University, Villejuif, France.,CellMedEx, Saint Maur Des Fossés, France
| | | | - Nassim Arouche
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - Bijan Bambai
- National Institute for Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France. .,Paris-Saclay University, Villejuif, France. .,CellMedEx, Saint Maur Des Fossés, France.
| |
Collapse
|
17
|
Quick S, Moss J, Rajani RM, Williams A. A Vessel for Change: Endothelial Dysfunction in Cerebral Small Vessel Disease. Trends Neurosci 2020; 44:289-305. [PMID: 33308877 DOI: 10.1016/j.tins.2020.11.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/24/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023]
Abstract
The blood vessels of the brain are lined with endothelial cells and it has been long known that these help to regulate blood flow to the brain. However, there is increasing evidence that these cells also interact with the surrounding brain tissue. These interactions change when the endothelial cells become dysfunctional and have an impact in diseases such as cerebral small vessel disease, the leading cause of vascular dementia. In this review, we focus on what endothelial dysfunction is, what causes it, how it leads to surrounding brain pathology, how researchers can investigate it with current models, and where this might lead in the future for dementia therapies.
Collapse
Affiliation(s)
- Sophie Quick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jonathan Moss
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
18
|
Vita SM, Redell JB, Maynard ME, Zhao J, Grill RJ, Dash PK, Grayson BE. P-glycoprotein Expression Is Upregulated in a Pre-Clinical Model of Traumatic Brain Injury. Neurotrauma Rep 2020; 1:207-217. [PMID: 33274346 PMCID: PMC7703495 DOI: 10.1089/neur.2020.0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Athletes participating in contact sports are at risk for sustaining repeat mild traumatic brain injury (rmTBI). Unfortunately, no pharmacological treatment to lessen the pathophysiology of brain injury has received U.S. Food and Drug Administration (FDA) approval. One hurdle to overcome for potential candidate agents to reach effective therapeutic concentrations in the brain is the blood-brain barrier (BBB). Adenosine triphosphate (ATP)-binding cassette (ABC) transporters, such as P-glycoprotein (Pgp), line the luminal membrane of the brain capillary endothelium facing the vascular space. Although these transporters serve to protect the central nervous system (CNS) from damage by effluxing neurotoxicants before they can reach the brain, they may also limit the accumulation of therapeutic drugs in the brain parenchyma. Thus, increased Pgp expression following brain injury may result in reduced brain availability of therapeutic agents. We therefore questioned if repeat concussive injury increases Pgp expression in the brain. To answer this question, we used a rodent model of repeat mild closed head injury (rmCHI) and examined the messenger RNA (mRN) and protein expression of both isoforms of rodent Pgp (Abcb1a and Abcb1b). Compared with sham-operated controls (n = 5), the mRNA levels of both Abcb1a and Abcb1b were found to be increased in the hippocampus at day 1 (n = 5) and at day 5 (n = 5) post-injury. Using a validated antibody, we show increased immunolabeling for Pgp in the dorsal cortex at day 5 and in the hippocampus at day 1 (n = 5) and at day 5 (n = 5) post-injury compared with sham controls (n = 6). Taken together, these results suggest that increased expression of Pgp after rmCHI may reduce the brain accumulation of therapeutic drugs that are Pgp substrates. It is plausible that including a Pgp inhibitor with a candidate therapeutic agent may be an effective approach to treat the pathophysiology of rmCHI.
Collapse
Affiliation(s)
- Sydney M Vita
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Mark E Maynard
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Jing Zhao
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Raymond J Grill
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, University of Texas McGovern Medical School, Houston, Texas, USA
| | - Bernadette E Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
19
|
Curtaz CJ, Schmitt C, Blecharz-Lang KG, Roewer N, Wöckel A, Burek M. Circulating MicroRNAs and Blood-Brain-Barrier Function in Breast Cancer Metastasis. Curr Pharm Des 2020; 26:1417-1427. [PMID: 32175838 PMCID: PMC7475800 DOI: 10.2174/1381612826666200316151720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/26/2020] [Indexed: 12/24/2022]
Abstract
Brain metastases are a major cause of death in breast cancer patients. A key event in the metastatic progression of breast cancer in the brain is the migration of cancer cells across the blood-brain barrier (BBB). The BBB is a natural barrier with specialized functions that protect the brain from harmful substances, including anti-tumor drugs. Extracellular vesicles (EVs) sequestered by cells are mediators of cell-cell communication. EVs carry cellular components, including microRNAs that affect the cellular processes of target cells. Here, we summarize the knowledge about microRNAs known to play a significant role in breast cancer and/or in the BBB function. In addition, we describe previously established in vitro BBB models, which are a useful tool for studying molecular mechanisms involved in the formation of brain metastases.
Collapse
Affiliation(s)
- Carolin J Curtaz
- Department of Gynecology and Obstetrics, University of Würzburg, Würzburg, Germany
| | - Constanze Schmitt
- Department of Anaesthesia and Critical Care, University of Würzburg, 97080 Würzburg, Germany
| | - Kinga G Blecharz-Lang
- Department of Experimental Neurosurgery, Charite - Universitätsmedizin, Berlin, Germany
| | - Norbert Roewer
- Department of Anaesthesia and Critical Care, University of Würzburg, 97080 Würzburg, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University of Würzburg, Würzburg, Germany
| | - Malgorzata Burek
- Department of Anaesthesia and Critical Care, University of Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
20
|
Nishihara H, Gastfriend BD, Soldati S, Perriot S, Mathias A, Sano Y, Shimizu F, Gosselet F, Kanda T, Palecek SP, Du Pasquier R, Shusta EV, Engelhardt B. Advancing human induced pluripotent stem cell-derived blood-brain barrier models for studying immune cell interactions. FASEB J 2020; 34:16693-16715. [PMID: 33124083 PMCID: PMC7686106 DOI: 10.1096/fj.202001507rr] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 12/16/2022]
Abstract
Human induced pluripotent stem cell (hiPSC)‐derived blood‐brain barrier (BBB) models established to date lack expression of key adhesion molecules involved in immune cell migration across the BBB in vivo. Here, we introduce the extended endothelial cell culture method (EECM), which differentiates hiPSC‐derived endothelial progenitor cells to brain microvascular endothelial cell (BMEC)‐like cells with good barrier properties and mature tight junctions. Importantly, EECM‐BMEC‐like cells exhibited constitutive cell surface expression of ICAM‐1, ICAM‐2, and E‐selectin. Pro‐inflammatory cytokine stimulation increased the cell surface expression of ICAM‐1 and induced cell surface expression of P‐selectin and VCAM‐1. Co‐culture of EECM‐BMEC‐like cells with hiPSC‐derived smooth muscle‐like cells or their conditioned medium further increased the induction of VCAM‐1. Functional expression of endothelial ICAM‐1 and VCAM‐1 was confirmed by T‐cell interaction with EECM‐BMEC‐like cells. Taken together, we introduce the first hiPSC‐derived BBB model that displays an adhesion molecule phenotype that is suitable for the study of immune cell interactions.
Collapse
Affiliation(s)
| | - Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Sylvain Perriot
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Amandine Mathias
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Fabien Gosselet
- Blood Brain Barrier Laboratory, University of Artois, Lens, France
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA.,Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | |
Collapse
|
21
|
Naserian S, Abdelgawad ME, Afshar Bakshloo M, Ha G, Arouche N, Cohen JL, Salomon BL, Uzan G. The TNF/TNFR2 signaling pathway is a key regulatory factor in endothelial progenitor cell immunosuppressive effect. Cell Commun Signal 2020; 18:94. [PMID: 32546175 PMCID: PMC7298859 DOI: 10.1186/s12964-020-00564-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Background Endothelial progenitor cells (EPCs) are non-differentiated endothelial cells (ECs) present in blood circulation that are involved in neo-vascularization and correction of damaged endothelial sites. Since EPCs from patients with vascular disorders are impaired and inefficient, allogenic sources from adult or cord blood are considered as good alternatives. However, due to the reaction of immune system against allogenic cells which usually lead to their elimination, we focused on the exact role of EPCs on immune cells, particularly, T cells which are the most important cells applied in immune rejection. TNFα is one of the main activators of EPCs that recognizes two distinct receptors. TNFR1 is expressed ubiquitously and its interaction with TNFα leads to differentiation and apoptosis, whereas, TNFR2 is expressed predominantly on ECs, immune cells and neural cells and is involved in cell survival and proliferation. Interestingly, it has been shown that different immunosuppressive cells express TNFR2 and this is directly related to their immunosuppressive efficiency. However, little is known about immunological profile and function of TNFR2 in EPCs. Methods Using different in-vitro combinations, we performed co-cultures of ECs and T cells to investigate the immunological effect of EPCs on T cells. We interrupted in the TNFα/TNFR2 axis either by blocking the receptor using TNFR2 antagonist or blocking the ligand using T cells derived from TNFα KO mice. Results We demonstrated that EPCs are able to suppress T cell proliferation and modulate them towards less pro-inflammatory and active phenotypes. Moreover, we showed that TNFα/TNFR2 immune-checkpoint pathway is critical in EPC immunomodulatory effect. Conclusions Our results reveal for the first time a mechanism that EPCs use to suppress immune cells, therefore, enabling them to form new immunosuppressive vessels. Furthermore, we have shown the importance of TNFα/TNFR2 axis in EPCs as an immune checkpoint pathway. We believe that targeting TNFR2 is especially crucial in cancer immune therapy since it controls two crucial aspects of tumor microenvironment: 1) Immunosuppression and 2) Angiogenesis. Video Abstract. (MP4 46355 kb)
Collapse
Affiliation(s)
- Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France. .,CellMedEx, Saint Maur Des Fossés, France. .,Paris-Saclay University, Villejuif, France.
| | - Mohamed Essameldin Abdelgawad
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Paris-Saclay University, Villejuif, France.,Biochemistry Division, Chemistry department, Faculty of Science, Helwan University, Cairo, Egypt
| | | | - Guillaume Ha
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
| | - Nassim Arouche
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France.,Paris-Saclay University, Villejuif, France
| | - José L Cohen
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France.,AP-HP, Hopital Henri Mondor, Centre d'investigation clinique biothérapie, F-94010, Creteil, France
| | - Benoît L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France. .,Paris-Saclay University, Villejuif, France.
| |
Collapse
|
22
|
Curtaz CJ, Schmitt C, Herbert SL, Feldheim J, Schlegel N, Gosselet F, Hagemann C, Roewer N, Meybohm P, Wöckel A, Burek M. Serum-derived factors of breast cancer patients with brain metastases alter permeability of a human blood-brain barrier model. Fluids Barriers CNS 2020; 17:31. [PMID: 32321535 PMCID: PMC7178982 DOI: 10.1186/s12987-020-00192-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/13/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The most threatening metastases in breast cancer are brain metastases, which correlate with a very poor overall survival, but also a limited quality of life. A key event for the metastatic progression of breast cancer into the brain is the migration of cancer cells across the blood-brain barrier (BBB). METHODS We adapted and validated the CD34+ cells-derived human in vitro BBB model (brain-like endothelial cells, BLECs) to analyse the effects of patient serum on BBB properties. We collected serum samples from healthy donors, breast cancer patients with primary cancer, and breast cancer patients with, bone, visceral or cerebral metastases. We analysed cytokine levels in these sera utilizing immunoassays and correlated them with clinical data. We used paracellular permeability measurements, immunofluorescence staining, Western blot and mRNA analysis to examine the effects of patient sera on the properties of BBB in vitro. RESULTS The BLECs cultured together with brain pericytes in transwells developed a tight monolayer with a correct localization of claudin-5 at the tight junctions (TJ). Several BBB marker proteins such as the TJ proteins claudin-5 and occludin, the glucose transporter GLUT-1 or the efflux pumps PG-P and BCRP were upregulated in these cultures. This was accompanied by a reduced paracellular permeability for fluorescein (400 Da). We then used this model for the treatment with the patient sera. Only the sera of breast cancer patients with cerebral metastases had significantly increased levels of the cytokines fractalkine (CX3CL1) and BCA-1 (CXCL13). The increased levels of fractalkine were associated with the estrogen/progesterone receptor status of the tumour. The treatment of BLECs with these sera selectively increased the expression of CXCL13 and TJ protein occludin. In addition, the permeability of fluorescein was increased after serum treatment. CONCLUSION We demonstrate that the CD34+ cell-derived human in vitro BBB model can be used as a tool to study the molecular mechanisms underlying cerebrovascular pathologies. We showed that serum from patients with cerebral metastases may affect the integrity of the BBB in vitro, associated with elevated concentrations of specific cytokines such as CX3CL1 and CXCL13.
Collapse
Affiliation(s)
- Carolin J Curtaz
- Department of Gynecology and Obstetrics, University of Würzburg, Würzburg, Germany
| | - Constanze Schmitt
- Department of Anaesthesia and Critical Care, University of Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | | | - Jonas Feldheim
- Department of Neurosurgery, Tumour Biology Laboratory, University of Würzburg, Würzburg, Germany
| | - Nicolas Schlegel
- Department of Surgery I, University of Würzburg, Würzburg, Germany
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory, Université d'Artois, UR, 2465, Lens, France
| | - Carsten Hagemann
- Department of Neurosurgery, Tumour Biology Laboratory, University of Würzburg, Würzburg, Germany
| | - Norbert Roewer
- Department of Anaesthesia and Critical Care, University of Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Patrick Meybohm
- Department of Anaesthesia and Critical Care, University of Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University of Würzburg, Würzburg, Germany
| | - Malgorzata Burek
- Department of Anaesthesia and Critical Care, University of Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany.
| |
Collapse
|
23
|
Bhalerao A, Sivandzade F, Archie SR, Chowdhury EA, Noorani B, Cucullo L. In vitro modeling of the neurovascular unit: advances in the field. Fluids Barriers CNS 2020; 17:22. [PMID: 32178700 PMCID: PMC7077137 DOI: 10.1186/s12987-020-00183-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) is a fundamental component of the central nervous system. Its functional and structural integrity is vital in maintaining the homeostasis of the brain microenvironment. On the other hand, the BBB is also a major hindering obstacle for the delivery of effective therapies to treat disorders of the Central Nervous System (CNS). Over time, various model systems have been established to simulate the complexities of the BBB. The development of realistic in vitro BBB models that accurately mimic the physiological characteristics of the brain microcapillaries in situ is of fundamental importance not only in CNS drug discovery but also in translational research. Successful modeling of the Neurovascular Unit (NVU) would provide an invaluable tool that would aid in dissecting out the pathological factors, mechanisms of action, and corresponding targets prodromal to the onset of CNS disorders. The field of BBB in vitro modeling has seen many fundamental changes in the last few years with the introduction of novel tools and methods to improve existing models and enable new ones. The development of CNS organoids, organ-on-chip, spheroids, 3D printed microfluidics, and other innovative technologies have the potential to advance the field of BBB and NVU modeling. Therefore, in this review, summarize the advances and progress in the design and application of functional in vitro BBB platforms with a focus on rapidly advancing technologies.
Collapse
Affiliation(s)
- Aditya Bhalerao
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Farzane Sivandzade
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA. .,Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
24
|
Raimondi I, Izzo L, Tunesi M, Comar M, Albani D, Giordano C. Organ-On-A-Chip in vitro Models of the Brain and the Blood-Brain Barrier and Their Value to Study the Microbiota-Gut-Brain Axis in Neurodegeneration. Front Bioeng Biotechnol 2020; 7:435. [PMID: 31998702 PMCID: PMC6965718 DOI: 10.3389/fbioe.2019.00435] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
We are accumulating evidence that intestinal microflora, collectively named gut microbiota, can alter brain pathophysiology, but researchers have just begun to discover the mechanisms of this bidirectional connection (often referred to as microbiota-gut-brain axis, MGBA). The most noticeable hypothesis for a pathological action of gut microbiota on the brain is based on microbial release of soluble neurotransmitters, hormones, immune molecules and neuroactive metabolites, but this complex scenario requires reliable and controllable tools for its causal demonstration. Thanks to three-dimensional (3D) cultures and microfluidics, engineered in vitro models could improve the scientific knowledge in this field, also from a therapeutic perspective. This review briefly retraces the main discoveries linking the activity of gut microbiota to prevalent brain neurodegenerative disorders, and then provides a deep insight into the state-of-the-art for in vitro modeling of the brain and the blood-brain barrier (BBB), two key players of the MGBA. Several brain and BBB microfluidic devices have already been developed to implement organ-on-a-chip solutions, but some limitations still exist. Future developments of organ-on-a-chip tools to model the MGBA will require an interdisciplinary approach and the synergy with cutting-edge technologies (for instance, bioprinting) to achieve multi-organ platforms and support basic research, also for the development of new therapies against neurodegenerative diseases.
Collapse
Affiliation(s)
- Ilaria Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Luca Izzo
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| | - Manola Comar
- SSD of Advanced Translational Microbiology, IRCCS “Burlo Garofolo”, Department of Medical Sciences (DMS), University of Trieste, Trieste, Italy
| | - Diego Albani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milan, Italy
| |
Collapse
|
25
|
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: 64] [Impact Index Per Article: 12.8] [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
|
26
|
Zhou Y, Shao A, Xu W, Wu H, Deng Y. Advance of Stem Cell Treatment for Traumatic Brain Injury. Front Cell Neurosci 2019; 13:301. [PMID: 31456663 PMCID: PMC6700304 DOI: 10.3389/fncel.2019.00301] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/19/2019] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of human mortality and morbidity, which can induce serious neurological damage. At present, clinical treatments for neurological dysfunction after TBI include hyperbaric oxygen, brain stimulation and behavioral therapy, but the therapeutic effect is not satisfactory. Recent studies have found that exogenous stem cells can migrate to damaged brain tissue, then participate in the repair of damaged brain tissue by further differentiation to replace damaged cells, while releasing anti-inflammatory factors and growth factors, thereby significantly improving neurological function. This article will mainly review the effects, deficiencies and related mechanisms of different types of stem cells in TBI.
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Ferreira L. What human blood-brain barrier models can tell us about BBB function and drug discovery? Expert Opin Drug Discov 2019; 14:1113-1123. [DOI: 10.1080/17460441.2019.1646722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
28
|
Williams IM, Wu JC. Generation of Endothelial Cells From Human Pluripotent Stem Cells. Arterioscler Thromb Vasc Biol 2019; 39:1317-1329. [PMID: 31242035 DOI: 10.1161/atvbaha.119.312265] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endothelial cells (ECs) are critical for several aspects of cardiovascular disease therapy, including vascular regeneration, personalized drug development, and tissue engineering. Human pluripotent stem cells (hPSCs) afford us with an unprecedented opportunity to produce virtually unlimited quantities of human ECs. In this review, we highlight key developments and outstanding challenges in our ability to derive ECs de novo from hPSCs. Furthermore, we consider strategies for recapitulating the vessel- and tissue-specific functional heterogeneity of ECs in vitro. Finally, we discuss ongoing attempts to utilize hPSC-derived ECs and their progenitors for various therapeutic applications. Continued progress in generating hPSC-derived ECs will profoundly enhance our ability to discover novel drug targets, revascularize ischemic tissues, and engineer clinically relevant tissue constructs. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Ian M Williams
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA
| | - Joseph C Wu
- From the Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, CA
| |
Collapse
|
29
|
Sato K. [Consideration for future in vitro BBB models - technical development to investigate the drug delivery to the CNS]. Nihon Yakurigaku Zasshi 2019; 152:287-294. [PMID: 30531099 DOI: 10.1254/fpj.152.287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Blood vessels in the central nervous system (CNS) limit the material exchange between blood and parenchyma by blood brain barrier (BBB). At present, no appropriate in vitro BBB models are available for the investigation whether or not the candidate compounds for new drugs could be delivered to the CNS. This causes huge difficulties of the development of CNS drugs and prediction of CNS adverse effects. In this review, I first outline the structures and functions of BBB, together with the parameters used for the quantification of BBB functions. I also introduce the history of in vitro BBB models used in the drug development so far, i.e., the transition from non-cell models to the models using primary culture of rodent cells, porcine, bovine, cell lines, etc. More recently, the application of human cells differentiated from human induced pluripotent stem cells and microfluidic engineering have already started. BBB is essential for the maintenance of brain homeostasis and the mechanisms of the BBB development will be clarified by reproducing functional BBB on the dish. The new in vitro models and the data may provide accurate prediction of drug delivery to the CNS and the improvement of the evaluation system for toxicity and safety, thereby leading to successful launch of new drugs on the market.
Collapse
|
30
|
Logan S, Arzua T, Canfield SG, Seminary ER, Sison SL, Ebert AD, Bai X. Studying Human Neurological Disorders Using Induced Pluripotent Stem Cells: From 2D Monolayer to 3D Organoid and Blood Brain Barrier Models. Compr Physiol 2019; 9:565-611. [PMID: 30873582 PMCID: PMC6705133 DOI: 10.1002/cphy.c180025] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurological disorders have emerged as a predominant healthcare concern in recent years due to their severe consequences on quality of life and prevalence throughout the world. Understanding the underlying mechanisms of these diseases and the interactions between different brain cell types is essential for the development of new therapeutics. Induced pluripotent stem cells (iPSCs) are invaluable tools for neurological disease modeling, as they have unlimited self-renewal and differentiation capacity. Mounting evidence shows: (i) various brain cells can be generated from iPSCs in two-dimensional (2D) monolayer cultures; and (ii) further advances in 3D culture systems have led to the differentiation of iPSCs into organoids with multiple brain cell types and specific brain regions. These 3D organoids have gained widespread attention as in vitro tools to recapitulate complex features of the brain, and (iii) complex interactions between iPSC-derived brain cell types can recapitulate physiological and pathological conditions of blood-brain barrier (BBB). As iPSCs can be generated from diverse patient populations, researchers have effectively applied 2D, 3D, and BBB models to recapitulate genetically complex neurological disorders and reveal novel insights into molecular and genetic mechanisms of neurological disorders. In this review, we describe recent progress in the generation of 2D, 3D, and BBB models from iPSCs and further discuss their limitations, advantages, and future ventures. This review also covers the current status of applications of 2D, 3D, and BBB models in drug screening, precision medicine, and modeling a wide range of neurological diseases (e.g., neurodegenerative diseases, neurodevelopmental disorders, brain injury, and neuropsychiatric disorders). © 2019 American Physiological Society. Compr Physiol 9:565-611, 2019.
Collapse
Affiliation(s)
- Sarah Logan
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Thiago Arzua
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Scott G. Canfield
- Department of Cellular & Integrative Physiology, IU School of Medicine-Terre Haute, Terre Haute, IN, USA
| | - Emily R. Seminary
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samantha L. Sison
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Allison D. Ebert
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
31
|
Waldau B. Using miniature brain implants in rodents for novel drug discovery. Expert Opin Drug Discov 2019; 14:379-386. [DOI: 10.1080/17460441.2019.1577816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Ben Waldau
- Department of Neurological Surgery, University of California, Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
32
|
Loisel F, Provost B, Guihaire J, Boulate D, Arouche N, Amsallem M, Arthur-Ataam J, Decante B, Dorfmüller P, Fadel E, Uzan G, Mercier O. Autologous endothelial progenitor cell therapy improves right ventricular function in a model of chronic thromboembolic pulmonary hypertension. J Thorac Cardiovasc Surg 2018; 157:655-666.e7. [PMID: 30669226 DOI: 10.1016/j.jtcvs.2018.08.083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 01/09/2023]
Abstract
BACKGROUND Right ventricular (RV) failure is the main prognostic factor in pulmonary hypertension, and ventricular capillary density (CD) has been reported to be a marker of RV maladaptive remodeling and failure. Our aim was to determine whether right intracoronary endothelial progenitor cell (EPC) infusion can improve RV function and CD in a piglet model of chronic thromboembolic pulmonary hypertension (CTEPH). METHODS We compared 3 groups: sham (n = 5), CTEPH (n = 6), and CTEPH with EPC infusion (CTEPH+EPC; n = 5). After EPC isolation from CTEPH+EPC piglet peripheral blood samples at 3 weeks, the CTEPH and sham groups underwent right intracoronary infusion of saline, and the CTEPH+EPC group received EPCs at 6 weeks. RV function, pulmonary hemodynamics, and myocardial morphometry were investigated in the animals at 10 weeks. RESULTS After EPC administration, the RV fractional area change increased from 32.75% (interquartile range [IQR], 29.5%-36.5%) to 39% (IQR, 37.25%-46.50%; P = .030). The CTEPH+EPC piglets had reduced cardiomyocyte surface areas (from 298.3 μm2 [IQR, 277.4-335.3 μm2] to 234.6 μm2 (IQR, 211.1-264.7 μm2; P = .017), and increased CD31 expression (from 3.12 [IQR, 1.27-5.09] to 7.14 [IQR, 5.56-8.41; P = .017). EPCs were found in the RV free wall at 4 and 24 hours after injection but not 4 weeks later. CONCLUSIONS Intracoronary infusion of EPC improved RV function and CD in a piglet model of CTEPH. This novel cell-based therapy might represent a promising RV-targeted treatment in patients with pulmonary hypertension.
Collapse
Affiliation(s)
- Fanny Loisel
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Inserm 1197 Research Unit, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Bastien Provost
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Julien Guihaire
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Department of Cardiac Surgery, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - David Boulate
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Nassim Arouche
- Inserm 1197 Research Unit, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Myriam Amsallem
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Jennifer Arthur-Ataam
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Benoît Decante
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Peter Dorfmüller
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Department of Pathology, Marie Lannelongue Hospital, Le Plessis Robinson, France
| | - Elie Fadel
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Paris-Sud University and Paris-Saclay University, School of Medicine, Kremlin-Bicêtre, France
| | - Georges Uzan
- Inserm 1197 Research Unit, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Olaf Mercier
- Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Univ Paris Sud, Paris-Saclay University, Le Plessis Robinson, France; Paris-Sud University and Paris-Saclay University, School of Medicine, Kremlin-Bicêtre, France.
| |
Collapse
|
33
|
Loisel F, Provost B, Haddad F, Guihaire J, Amsallem M, Vrtovec B, Fadel E, Uzan G, Mercier O. Stem cell therapy targeting the right ventricle in pulmonary arterial hypertension: is it a potential avenue of therapy? Pulm Circ 2018; 8:2045893218755979. [PMID: 29480154 PMCID: PMC5844533 DOI: 10.1177/2045893218755979] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an incurable disease characterized by an increase in pulmonary arterial pressure due to pathological changes to the pulmonary vascular bed. As a result, the right ventricle (RV) is subject to an increased afterload and undergoes multiple changes, including a decrease in capillary density. All of these dysfunctions lead to RV failure. A number of studies have shown that RV function is one of the main prognostic factors for PAH patients. Many stem cell therapies targeting the left ventricle are currently undergoing development. The promising results observed in animal models have led to clinical trials that have shown an improvement of cardiac function. In contrast to left heart disease, stem cell therapy applied to the RV has remained poorly studied, even though it too may provide a therapeutic benefit. In this review, we discuss stem cell therapy as a treatment for RV failure in PAH. We provide an overview of the results of preclinical and clinical studies for RV cell therapies. Although a large number of studies have targeted the pulmonary circulation rather than the RV directly, there are nonetheless encouraging results in the literature that indicate that cell therapies may have a direct beneficial effect on RV function. This cell therapy strategy may therefore hold great promise and warrants further studies in PAH patients.
Collapse
Affiliation(s)
- Fanny Loisel
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,2 Inserm 1197 Research Unit, Universite Paris Sud, Paris-Saclay University, Villejuif, France
| | - Bastien Provost
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - François Haddad
- 3 Cardiovascular Medicine, Stanford Hospital, Stanford University, CA, USA
| | - Julien Guihaire
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Myriam Amsallem
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Bojan Vrtovec
- 4 Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Elie Fadel
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,5 Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| | - Georges Uzan
- 2 Inserm 1197 Research Unit, Universite Paris Sud, Paris-Saclay University, Villejuif, France
| | - Olaf Mercier
- 1 36705 Research and Innovation Unit, Inserm UMR-S 999, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France.,5 Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, Universite Paris Sud, Paris-Saclay University, Le Plessis Robinson, France
| |
Collapse
|
34
|
Neurovascular dysfunction in dementia - human cellular models and molecular mechanisms. Clin Sci (Lond) 2018; 132:399-418. [PMID: 29444850 DOI: 10.1042/cs20160720] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/15/2018] [Accepted: 01/19/2018] [Indexed: 02/08/2023]
Abstract
From the earliest stages of development, when cerebral angiogenesis and neurogenesis are entwined, to the end of life, the interplay between vascular and neural systems of the brain is critical in health and disease. Cerebral microvascular endothelial cells constitute the blood-brain barrier and in concert with pericytes or smooth muscle cells, glia and neurons, integrate into a functional neurovascular unit (NVU). This multicellular NVU maintains homoeostasis of the brain's microenvironment by restricting the entry of systemic pathogens and neurotoxins as well as meeting the metabolic demands of neural activity. Recent evidence of cerebral microvascular pathologies in vascular diseases and dementia, including Alzheimer's disease, has challenged the notion that vascular events are merely the consequence of neuronal pathology. This review focuses on molecular mechanisms of neurovascular dysfunction in dementia and outlines currently employed in vitro models to decode such mechanisms. Deciphering neurovascular crosstalk is likely to be more important in understanding the molecular mechanisms of disease than previously anticipated and may offer novel therapeutic opportunities for dementia and related conditions.
Collapse
|
35
|
Ribecco-Lutkiewicz M, Sodja C, Haukenfrers J, Haqqani AS, Ly D, Zachar P, Baumann E, Ball M, Huang J, Rukhlova M, Martina M, Liu Q, Stanimirovic D, Jezierski A, Bani-Yaghoub M. A novel human induced pluripotent stem cell blood-brain barrier model: Applicability to study antibody-triggered receptor-mediated transcytosis. Sci Rep 2018; 8:1873. [PMID: 29382846 PMCID: PMC5789839 DOI: 10.1038/s41598-018-19522-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 12/27/2017] [Indexed: 12/21/2022] Open
Abstract
We have developed a renewable, scalable and transgene free human blood-brain barrier model, composed of brain endothelial cells (BECs), generated from human amniotic fluid derived induced pluripotent stem cells (AF-iPSC), which can also give rise to syngeneic neural cells of the neurovascular unit. These AF-iPSC-derived BECs (i-BEC) exhibited high transendothelial electrical resistance (up to 1500 Ω cm2) inducible by astrocyte-derived molecular cues and retinoic acid treatment, polarized expression of functional efflux transporters and receptor mediated transcytosis triggered by antibodies against specific receptors. In vitro human BBB models enable pre-clinical screening of central nervous system (CNS)-targeting drugs and are of particular importance for assessing species-specific/selective transport mechanisms. This i-BEC human BBB model discriminates species-selective antibody- mediated transcytosis mechanisms, is predictive of in vivo CNS exposure of rodent cross-reactive antibodies and can be implemented into pre-clinical CNS drug discovery and development processes.
Collapse
Affiliation(s)
- Maria Ribecco-Lutkiewicz
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Caroline Sodja
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Julie Haukenfrers
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Arsalan S Haqqani
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Dao Ly
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Peter Zachar
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Ewa Baumann
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Marguerite Ball
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Jez Huang
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Marina Rukhlova
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Marzia Martina
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Qing Liu
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Danica Stanimirovic
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| | - Anna Jezierski
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada.
| | - Mahmud Bani-Yaghoub
- Human Health Therapeutics Portfolio, National Research Council of Canada, Ottawa, ON, K1A 0R6, Canada
| |
Collapse
|
36
|
Gonzalez DM, Gregory J, Brennand KJ. The Importance of Non-neuronal Cell Types in hiPSC-Based Disease Modeling and Drug Screening. Front Cell Dev Biol 2017; 5:117. [PMID: 29312938 PMCID: PMC5742170 DOI: 10.3389/fcell.2017.00117] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
Current applications of human induced pluripotent stem cell (hiPSC) technologies in patient-specific models of neurodegenerative and neuropsychiatric disorders tend to focus on neuronal phenotypes. Here, we review recent efforts toward advancing hiPSCs toward non-neuronal cell types of the central nervous system (CNS) and highlight their potential use for the development of more complex in vitro models of neurodevelopment and disease. We present evidence from previous works in both rodents and humans of the importance of these cell types (oligodendrocytes, microglia, astrocytes) in neurological disease and highlight new hiPSC-based models that have sought to explore these relationships in vitro. Lastly, we summarize efforts toward conducting high-throughput screening experiments with hiPSCs and propose methods by which new screening platforms could be designed to better capture complex relationships between neural cell populations in health and disease.
Collapse
Affiliation(s)
- David M Gonzalez
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Developmental and Stem Cell Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jill Gregory
- Instructional Technology Group, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristen J Brennand
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
37
|
Wilson HK, Faubion MG, Hjortness MK, Palecek SP, Shusta EV. Cryopreservation of Brain Endothelial Cells Derived from Human Induced Pluripotent Stem Cells Is Enhanced by Rho-Associated Coiled Coil-Containing Kinase Inhibition. Tissue Eng Part C Methods 2017; 22:1085-1094. [PMID: 27846787 DOI: 10.1089/ten.tec.2016.0345] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The blood-brain barrier (BBB) maintains brain homeostasis but also presents a major obstacle to brain drug delivery. Brain microvascular endothelial cells (BMECs) form the principal barrier and therefore represent the major cellular component of in vitro BBB models. Such models are often used for mechanistic studies of the BBB in health and disease and for drug screening. Recently, human induced pluripotent stem cells (iPSCs) have emerged as a new source for generating BMEC-like cells for use in in vitro human BBB studies. However, the inability to cryopreserve iPSC-BMECs has impeded implementation of this model by requiring a fresh differentiation to generate cells for each experiment. Cryopreservation of differentiated iPSC-BMECs would have a number of distinct advantages, including enabling production of larger scale lots, decreasing lead time to generate purified iPSC-BMEC cultures, and facilitating use of iPSC-BMECs in large-scale screening. In this study, we demonstrate that iPSC-BMECs can be successfully cryopreserved at multiple differentiation stages. Cryopreserved iPSC-BMECs retain high viability, express standard endothelial and BBB markers, and reach a high transendothelial electrical resistance (TEER) of ∼3000 Ω·cm2, equivalent to nonfrozen controls. Rho-associated coiled coil-containing kinase (ROCK) inhibitor Y-27632 substantially increased survival and attachment of cryopreserved iPSC-BMECs, as well as stabilized TEER above 800 Ω·cm2 out to 7 days post-thaw. Overall, cryopreservation will ease handling and storage of high-quality iPSC-BMECs, reducing a key barrier to greater implementation of these cells in modeling the human BBB.
Collapse
Affiliation(s)
- Hannah K Wilson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison , Madison, Wisconsin
| | - Madeline G Faubion
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison , Madison, Wisconsin
| | - Michael K Hjortness
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison , Madison, Wisconsin
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison , Madison, Wisconsin
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison , Madison, Wisconsin
| |
Collapse
|
38
|
Qian T, Maguire SE, Canfield SG, Bao X, Olson WR, Shusta EV, Palecek SP. Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells. SCIENCE ADVANCES 2017; 3:e1701679. [PMID: 29134197 PMCID: PMC5677350 DOI: 10.1126/sciadv.1701679] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/11/2017] [Indexed: 05/18/2023]
Abstract
The blood-brain barrier (BBB) is composed of specialized endothelial cells that are critical to neurological health. A key tool for understanding human BBB development and its role in neurological disease is a reliable and scalable source of functional brain microvascular endothelial cells (BMECs). Human pluripotent stem cells (hPSCs) can theoretically generate unlimited quantities of any cell lineage in vitro, including BMECs, for disease modeling, drug screening, and cell-based therapies. We demonstrate a facile, chemically defined method to differentiate hPSCs to BMECs in a developmentally relevant progression via small-molecule activation of key signaling pathways. hPSCs are first induced to mesoderm commitment by activating canonical Wnt signaling. Next, these mesoderm precursors progress to endothelial progenitors, and treatment with retinoic acid leads to acquisition of BBB-specific markers and phenotypes. hPSC-derived BMECs generated via this protocol exhibit endothelial properties, including tube formation and low-density lipoprotein uptake, as well as efflux transporter activities characteristic of BMECs. Notably, these cells exhibit high transendothelial electrical resistance above 3000 ohm·cm2. These hPSC-derived BMECs serve as a robust human in vitro BBB model that can be used to study brain disease and inform therapeutic development.
Collapse
Affiliation(s)
- Tongcheng Qian
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Shaenah E. Maguire
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Scott G. Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Xiaoping Bao
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - William R. Olson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Eric V. Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
39
|
Ferratge S, Boyer J, Arouch N, Chevalier F, Uzan G. Circulating endothelial progenitors in vascular repair. Biomed Mater Eng 2017; 28:S65-S74. [PMID: 28372279 DOI: 10.3233/bme-171625] [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: 12/22/2022]
Abstract
Endothelial Colony Forming Cells (ECFCs) are obtained in culture from Circulating Endothelial Progenitor Cells. They display all characteristics of endothelial cells and they display stem cells features. Cord blood-derived ECFCs (CB-ECFCs) have a high clonogenic and proliferative potentials, and exhibit vascular repair capabilities useful for the treatment of ischemic diseases. However, the link between immaturity and functional properties of CB-ECFCs is still poorly defined. We showed that these cells have a high clonogenic potential and are capable to be efficiently reprogrammed into induced pluripotent stem cells. Moreover, we analyzed the expression of a broad panel of genes involved in embryonic stem cell properties. We define a novel stem cell transcriptional signature for CB-ECFCs fora better characterization and stratification according to their stem cell profile. We then improved the yield of CB-ECFC production for obtaining cells more functional in fewer passages. We used Glycosaminoglycans (GAG), components from the extracellular matrix which potentiate heparin binding growth factor activities. GAG mimetics were designed, having the capacity to increase the yield of ECFC during the isolation process, to increase the number of colonies, improve adhesion, proliferation, migration and self-renewal. GAG mimetics have thus great interest for vascular regeneration in combination with ECFC. Our results show that CB-ECFC are immature cells harboring specific functions such as formation of colonies, proliferation and formation of vascular structures in vitro and in vivo.
Collapse
Affiliation(s)
- S Ferratge
- Inserm U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94807 Villejuif Cedex, France
| | - J Boyer
- Inserm U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94807 Villejuif Cedex, France
| | - N Arouch
- Inserm U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94807 Villejuif Cedex, France
| | - F Chevalier
- Inserm U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94807 Villejuif Cedex, France
| | - G Uzan
- Inserm U1197, Hôpital Paul Brousse, Bâtiment Lavoisier, 12-14 avenue Paul Vaillant Couturier, 94807 Villejuif Cedex, France
| |
Collapse
|
40
|
Peters EB. Endothelial Progenitor Cells for the Vascularization of Engineered Tissues. TISSUE ENGINEERING PART B-REVIEWS 2017; 24:1-24. [PMID: 28548628 DOI: 10.1089/ten.teb.2017.0127] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Self-assembled microvasculature from cocultures of endothelial cells (ECs) and stromal cells has significantly advanced efforts to vascularize engineered tissues by enhancing perfusion rates in vivo and producing investigative platforms for microvascular morphogenesis in vitro. However, to clinically translate prevascularized constructs, the issue of EC source must be resolved. Endothelial progenitor cells (EPCs) can be noninvasively supplied from the recipient through adult peripheral and umbilical cord blood, as well as derived from induced pluripotent stem cells, alleviating antigenicity issues. EPCs can also differentiate into all tissue endothelium, and have demonstrated potential for therapeutic vascularization. Yet, EPCs are not the standard EC choice to vascularize tissue constructs in vitro. Possible reasons include unresolved issues with EPC identity and characterization, as well as uncertainty in the selection of coculture, scaffold, and culture media combinations that promote EPC microvessel formation. This review addresses these issues through a summary of EPC vascular biology and the effects of tissue engineering design parameters upon EPC microvessel formation. Also included are perspectives to integrate EPCs with emerging technologies to produce functional, organotypic vascularized tissues.
Collapse
Affiliation(s)
- Erica B Peters
- Department of Chemical and Biological Engineering, University of Colorado , Boulder, Colorado
| |
Collapse
|
41
|
Lauschke K, Frederiksen L, Hall VJ. Paving the Way Toward Complex Blood-Brain Barrier Models Using Pluripotent Stem Cells. Stem Cells Dev 2017; 26:857-874. [PMID: 28398169 DOI: 10.1089/scd.2017.0003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A tissue with great need to be modeled in vitro is the blood-brain barrier (BBB). The BBB is a tight barrier that covers all blood vessels in the brain and separates the brain microenvironment from the blood system. It consists of three cell types [neurovascular unit (NVU)] that contribute to the unique tightness and selective permeability of the BBB and has been shown to be disrupted in many diseases and brain disorders, such as vascular dementia, stroke, multiple sclerosis, and Alzheimer's disease. Given the progress that pluripotent stem cells (PSCs) have made in the past two decades, it is now possible to produce many cell types from the BBB and even partially recapitulate this complex tissue in vitro. In this review, we summarize the most recent developments in PSC differentiation and modeling of the BBB. We also suggest how patient-specific human-induced PSCs could be used to model BBB dysfunction in the future. Lastly, we provide perspectives on how to improve production of the BBB in vitro, for example by improving pericyte differentiation protocols and by better modeling the NVU in the dish.
Collapse
Affiliation(s)
- Karin Lauschke
- 1 National Food Institute, Technical University of Denmark , Kongens Lyngby, Denmark
- 2 Department of Micro- and Nanotechnology, Technical University of Denmark , Kongens Lyngby, Denmark
| | - Lise Frederiksen
- 3 Faculty of Health and Medical Sciences, University of Copenhagen , København N, Denmark
| | - Vanessa Jane Hall
- 4 Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen , Frederiksberg C, Denmark
| |
Collapse
|
42
|
Abstract
The blood-brain barrier (BBB) is located at the brain microvessel level and isolates the brain from the whole body, thus restricting molecule and cell exchanges between cerebral and peripheral compartments. In order to better decipher and understand the BBB physiology and development, and to investigate transport mechanism and toxicity of neuropharmaceuticals, several in vitro BBB models have been developed using animal or human cells, primary or immortalized cells. The aim of this review is to explain to the reader the major criteria required for a pertinent in vitro BBB model and to briefly expose the different models currently available with their characteristics with a special focus on the static models.
Collapse
Affiliation(s)
- Fabien Gosselet
- Université Artois, EA 2465, laboratoire de la Barrière Hémato-Encéphalique (LBHE), rue Jean Souvraz, SP18, F-62300 Lens, France
| |
Collapse
|
43
|
Ferratge S, Ha G, Carpentier G, Arouche N, Bascetin R, Muller L, Germain S, Uzan G. Initial clonogenic potential of human endothelial progenitor cells is predictive of their further properties and establishes a functional hierarchy related to immaturity. Stem Cell Res 2017; 21:148-159. [PMID: 28499264 DOI: 10.1016/j.scr.2017.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 01/24/2017] [Accepted: 04/14/2017] [Indexed: 12/13/2022] Open
Abstract
Endothelial progenitor cells (EPCs) generate in vitro Endothelial Colony Forming Cells (ECFCs) combining features of endothelial and stem/progenitor cells. Their angiogenic properties confer them a therapeutic potential for treating ischemic lesions. They may be isolated from umbilical cord blood (CB-ECFCs) or peripheral adult blood (AB-ECFCs). It is generally accepted that CB-ECFCs are more clonogenic, proliferative and angiogenic than AB-ECFCs. Nevertheless, only a few studies have focused on the functional heterogeneity of CB-ECFCs from different individuals. Moreover, AB-ECFC loss of function is yet to be precisely described. We have focused on these two issues that are critical for clinical perspectives. The detailed clonogenic profile of CB-ECFCs and AB-ECFCs was obtained and revealed a high inter individual heterogeneity and the absence of correlation with age. Most CB-ECFCs yielded initial colonies and had functional properties similar to those of AB-ECFCs. Conversely, a high clonogenicity was associated with an enhanced proliferative and angiogenic potential and stemness gene overexpression, confirming that immaturity, lost by AB-ECFCs, was a prerequisite to functionality. We thus demonstrated the importance of selecting CB-ECFCs according to specific criteria, and we propose using the initial clonogenicity as a relevant marker of their potential efficacy on vascular repair.
Collapse
Affiliation(s)
| | - Guillaume Ha
- INSERM U1197, Hôpital Paul Brousse, Villejuif, France
| | - Gilles Carpentier
- ERL CNRS 9215, Laboratoire CRRET, Université Paris Est Créteil, Faculté des Sciences et Technologies, Créteil, France
| | | | - Rümeyza Bascetin
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France; Inserm U1050, Paris, France; CNRS UMRS 7241, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France; Inserm U1050, Paris, France; CNRS UMRS 7241, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France; Inserm U1050, Paris, France; CNRS UMRS 7241, Paris, France
| | - Georges Uzan
- INSERM U1197, Hôpital Paul Brousse, Villejuif, France.
| |
Collapse
|
44
|
Appelt-Menzel A, Cubukova A, Günther K, Edenhofer F, Piontek J, Krause G, Stüber T, Walles H, Neuhaus W, Metzger M. Establishment of a Human Blood-Brain Barrier Co-culture Model Mimicking the Neurovascular Unit Using Induced Pluri- and Multipotent Stem Cells. Stem Cell Reports 2017; 8:894-906. [PMID: 28344002 PMCID: PMC5390136 DOI: 10.1016/j.stemcr.2017.02.021] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 11/28/2022] Open
Abstract
In vitro models of the human blood-brain barrier (BBB) are highly desirable for drug development. This study aims to analyze a set of ten different BBB culture models based on primary cells, human induced pluripotent stem cells (hiPSCs), and multipotent fetal neural stem cells (fNSCs). We systematically investigated the impact of astrocytes, pericytes, and NSCs on hiPSC-derived BBB endothelial cell function and gene expression. The quadruple culture models, based on these four cell types, achieved BBB characteristics including transendothelial electrical resistance (TEER) up to 2,500 Ω cm2 and distinct upregulation of typical BBB genes. A complex in vivo-like tight junction (TJ) network was detected by freeze-fracture and transmission electron microscopy. Treatment with claudin-specific TJ modulators caused TEER decrease, confirming the relevant role of claudin subtypes for paracellular tightness. Drug permeability tests with reference substances were performed and confirmed the suitability of the models for drug transport studies. Establishment of a standardized human BBB co-culture model based on hiPSCs and fNSCs Reflection of physiological BBB integrity and expression of relevant transporters/TJs Confirmation of TJ network functionality by claudin-specific TJ modulators Validation of physiological transcellular model tightness by permeability studies
Collapse
Affiliation(s)
- Antje Appelt-Menzel
- University Hospital Würzburg, Chair Tissue Engineering and Regenerative Medicine, 97070 Würzburg, Germany; Translational Center Würzburg "Regenerative Therapies for Oncology and Musculoskeletal Diseases", Branch of Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 97070 Würzburg, Germany
| | - Alevtina Cubukova
- Translational Center Würzburg "Regenerative Therapies for Oncology and Musculoskeletal Diseases", Branch of Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 97070 Würzburg, Germany
| | - Katharina Günther
- Julius-Maximilians-University Würzburg, Institute of Anatomy and Cell Biology, Stem Cell and Regenerative Medicine Group, 97070 Würzburg, Germany
| | - Frank Edenhofer
- Julius-Maximilians-University Würzburg, Institute of Anatomy and Cell Biology, Stem Cell and Regenerative Medicine Group, 97070 Würzburg, Germany; Leopold-Franzens-University Innsbruck, Institute of Molecular Biology & CMBI, Department Genomics, Stem Cell Biology & Regenerative Medicine, 6020 Innsbruck, Austria
| | - Jörg Piontek
- Charité Universitätsmedizin Berlin, Clinical Physiology & Nutritional Medicine, Department of Gastroenterology, Rheumatology & Infectious Diseases, 12203 Berlin, Germany
| | - Gerd Krause
- Leibniz Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Tanja Stüber
- University Hospital Würzburg, Women's Hospital and Polyclinic, 97080 Würzburg, Germany
| | - Heike Walles
- University Hospital Würzburg, Chair Tissue Engineering and Regenerative Medicine, 97070 Würzburg, Germany; Translational Center Würzburg "Regenerative Therapies for Oncology and Musculoskeletal Diseases", Branch of Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 97070 Würzburg, Germany
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbH, Competence Center Health and Bioresources, Competence Unit Molecular Diagnostics, 1190 Vienna, Austria
| | - Marco Metzger
- University Hospital Würzburg, Chair Tissue Engineering and Regenerative Medicine, 97070 Würzburg, Germany; Translational Center Würzburg "Regenerative Therapies for Oncology and Musculoskeletal Diseases", Branch of Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 97070 Würzburg, Germany.
| |
Collapse
|
45
|
Stem Cells as a Promising Tool for the Restoration of Brain Neurovascular Unit and Angiogenic Orientation. Mol Neurobiol 2016; 54:7689-7705. [DOI: 10.1007/s12035-016-0286-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 11/02/2016] [Indexed: 02/07/2023]
|
46
|
Kaisar MA, Sajja RK, Prasad S, Abhyankar VV, Liles T, Cucullo L. New experimental models of the blood-brain barrier for CNS drug discovery. Expert Opin Drug Discov 2016; 12:89-103. [PMID: 27782770 DOI: 10.1080/17460441.2017.1253676] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a dynamic biological interface which actively controls the passage of substances between the blood and the central nervous system (CNS). From a biological and functional standpoint, the BBB plays a crucial role in maintaining brain homeostasis inasmuch that deterioration of BBB functions are prodromal to many CNS disorders. Conversely, the BBB hinders the delivery of drugs targeting the brain to treat a variety of neurological diseases. Area covered: This article reviews recent technological improvements and innovation in the field of BBB modeling including static and dynamic cell-based platforms, microfluidic systems and the use of stem cells and 3D printing technologies. Additionally, the authors laid out a roadmap for the integration of microfluidics and stem cell biology as a holistic approach for the development of novel in vitro BBB platforms. Expert opinion: Development of effective CNS drugs has been hindered by the lack of reliable strategies to mimic the BBB and cerebrovascular impairments in vitro. Technological advancements in BBB modeling have fostered the development of highly integrative and quasi- physiological in vitro platforms to support the process of drug discovery. These advanced in vitro tools are likely to further current understanding of the cerebrovascular modulatory mechanisms.
Collapse
Affiliation(s)
- Mohammad A Kaisar
- a Department of Pharmaceutical Sciences , Texas Tech University Health Sciences Center , Amarillo , TX , USA
| | - Ravi K Sajja
- a Department of Pharmaceutical Sciences , Texas Tech University Health Sciences Center , Amarillo , TX , USA
| | - Shikha Prasad
- a Department of Pharmaceutical Sciences , Texas Tech University Health Sciences Center , Amarillo , TX , USA
| | - Vinay V Abhyankar
- c Biological Microsystems Division at The University of Texas at Arlington Research Institute , Fort Worth , TX , USA
| | - Taylor Liles
- a Department of Pharmaceutical Sciences , Texas Tech University Health Sciences Center , Amarillo , TX , USA
| | - Luca Cucullo
- a Department of Pharmaceutical Sciences , Texas Tech University Health Sciences Center , Amarillo , TX , USA.,b Center for Blood Brain Barrier Research , Texas Tech University Health Sciences Center , Amarillo , TX , USA
| |
Collapse
|
47
|
Biemans EALM, Jäkel L, de Waal RMW, Kuiperij HB, Verbeek MM. Limitations of the hCMEC/D3 cell line as a model for Aβ clearance by the human blood-brain barrier. J Neurosci Res 2016; 95:1513-1522. [PMID: 27726164 PMCID: PMC5484315 DOI: 10.1002/jnr.23964] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease and cerebral amyloid angiopathy are characterized by accumulation of amyloid-β (Aβ) at the cerebrovasculature due to decreased clearance at the blood-brain barrier (BBB). However, the exact mechanism of Aβ clearance across this barrier has not been fully elucidated. The hCMEC/D3 cell line has been characterized as a valid model for the BBB. In this study we evaluated the use of this model to study Aβ clearance across the BBB, with an emphasis on brain-to-blood directional permeability. Barrier integrity of hCMEC/D3 monolayers was confirmed for large molecules in both the apical to basolateral and the reverse direction. However, permeability for smaller molecules was substantially higher, especially in basolateral to apical direction, and barrier formation for Aβ was completely absent in this direction. In addition, hCMEC/D3 cells failed to develop a high TEER, possibly caused by incomplete formation of tight junctions. We conclude that the hCMEC/D3 model has several limitations to study the cerebral clearance of Aβ. Therefore, the model needs further characterization before this cell system can be generally applied as a model to study cerebral Aβ clearance. © 2016 The Authors Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elisanne A L M Biemans
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Lieke Jäkel
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Robert M W de Waal
- Radboud University Medical Center, Department of Pathology, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| |
Collapse
|
48
|
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: 4.8] [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]
|
49
|
Hajjar I, Goldstein FC, Waller EK, Moss LD, Quyyumi A. Circulating Progenitor Cells is Linked to Cognitive Decline in Healthy Adults. Am J Med Sci 2016; 351:147-52. [PMID: 26897269 DOI: 10.1016/j.amjms.2015.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/14/2015] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Cognitive and cardiovascular disorders share many risk factors. Higher bone-marrow derived progenitor cells (PC) in blood are associated with lower rates of cardiovascular events but the association of PC with cognitive function is unclear. The objective of this study was to assess the association between PC and cognition in a sample of healthy adults enrolled in a cohort study. MATERIALS AND METHODS A random sample of employees at Emory University and Georgia Institute of Technology were followed for 4 years and underwent yearly vascular and cognitive assessment (N = 430, mean age = 49.2 years, 70% women, and 27% African-American). Cognition was assessed using computerized versions of 15 cognitive tests and principal component analysis was used for deriving cognitive scores: executive function, memory and working memory. PC were defined as mononuclear cells with specific surface markers (7 phenotypes). Decreased cognition in a domain was defined as performing below the lowest quartile for the corresponding domain at baseline. Generalized estimating equations were used to investigate associations between PC and cognition. RESULTS Higher PC levels at baseline were associated with lower risk of cognitive decline in the executive and working memory domains during the follow-up period (P < 0.002 for all PC phenotypes). Further, the degree of decline in PC over the follow-up period was correlated with a corresponding decline in performances in all 3 cognitive domains over the same period (All P < 0.002). CONCLUSION Lower PC and greater yearly declines in PC are associated with greater cognitive decline. These findings suggest the role for PC in neurocognitive aging.
Collapse
Affiliation(s)
- Ihab Hajjar
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Emory University, Atlanta, Georgia.
| | | | - Edmund K Waller
- Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Lauren D Moss
- Program Management Associate, Sarah Cannon Research Institute, Nashville, Tennessee
| | - Arshed Quyyumi
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
50
|
Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Förster C, Galla HJ, Romero IA, Shusta EV, Stebbins MJ, Vandenhaute E, Weksler B, Brodin B. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab 2016; 36:862-90. [PMID: 26868179 PMCID: PMC4853841 DOI: 10.1177/0271678x16630991] [Citation(s) in RCA: 525] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
The endothelial cells lining the brain capillaries separate the blood from the brain parenchyma. The endothelial monolayer of the brain capillaries serves both as a crucial interface for exchange of nutrients, gases, and metabolites between blood and brain, and as a barrier for neurotoxic components of plasma and xenobiotics. This "blood-brain barrier" function is a major hindrance for drug uptake into the brain parenchyma. Cell culture models, based on either primary cells or immortalized brain endothelial cell lines, have been developed, in order to facilitate in vitro studies of drug transport to the brain and studies of endothelial cell biology and pathophysiology. In this review, we aim to give an overview of established in vitro blood-brain barrier models with a focus on their validation regarding a set of well-established blood-brain barrier characteristics. As an ideal cell culture model of the blood-brain barrier is yet to be developed, we also aim to give an overview of the advantages and drawbacks of the different models described.
Collapse
Affiliation(s)
- Hans C Helms
- Department of Pharmacy, University of Copenhagen, Denmark
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, UK
| | - Malgorzata Burek
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | | | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, HAS, Szeged, Hungary
| | - Carola Förster
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | - Hans J Galla
- Institute of Biochemistry, University of Muenster, Germany
| | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | | | - Babette Weksler
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, NY, USA
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Denmark
| |
Collapse
|