1
|
Cao X, Ma R, Wang Y, Huang Y, You K, Zhang L, Li H, Feng G, Chen T, Wang D, Sun K, Fang H, Shen X. Paeoniflorin protects the vascular endothelial barrier in mice with sepsis by activating RXRα signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156384. [PMID: 39826282 DOI: 10.1016/j.phymed.2025.156384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Sepsis is a life-threatening condition characterized by organ dysfunction resulting from the body's aberrant response to infection. A primary indicator of early sepsis is vascular leakage due to endothelial injury. The immunomodulatory effects of paeoniflorin are well established. However, its effect on vascular endothelial injury in sepsis remains to be verified. METHODS The sepsis model was established by cecal ligation and puncture (CLP), along with simultaneous administration of paeoniflorin. The therapeutic effectiveness of paeoniflorin was evaluated by assessing the survival rate, the bacterial load in blood and the histopathological lung tissue injury. The pulmonary vascular endothelial barrier integrity was assessed using immunofluorescence, western blot, Evans blue dye, and qPCR. Human umbilical vein endothelial cells (HUVECs) were used for in vitro validation and exploration of the underlying mechanisms. RESULTS The CLP mice exhibited significant damage to pulmonary tissue and breakdown of endothelial barrier. Administration of paeoniflorin markedly improved survival rates, mitigated lung injury, and preserved the integrity of the pulmonary vascular endothelial barrier in CLP mice which was confirmed by in vitro experiments. Pharmacological mechanism studies showed that the protective effects of paeoniflorin on the vascular endothelium was achieved through activation of RXRα signaling, which could be reversed by RXRα knockdown. CONCLUSION Our experiments demonstrates the protective effect of paeoniflorin on the vascular endothelial barrier through activation of the RXRα, thereby offering potential therapeutic options for sepsis treatment. We also identified RXRα as a novel transcription factor for VE-cadherin, providing a potential new intervention target for vascular endothelial barrier damage in sepsis.
Collapse
Affiliation(s)
- Xinyue Cao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Ruihua Ma
- Department of Anesthesiology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, PR China, 215125
| | - Yirui Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Yuran Huang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Keyuan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Lijie Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Haidong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Guize Feng
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Tongqing Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Dong Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China
| | - Keyu Sun
- Department of Emergency, Minhang Hospital, Fudan University, Shanghai, PR China, 201199.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, PR China, 200032; Department of Anesthesiology, Shanghai Geriatic Medical Center, Shanghai, PR China, 201104; Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, PR China, 201199.
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, PR China.
| |
Collapse
|
2
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2024. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
3
|
Ding Y, Palecek SP, Shusta EV. iPSC-derived blood-brain barrier modeling reveals APOE isoform-dependent interactions with amyloid beta. Fluids Barriers CNS 2024; 21:79. [PMID: 39394110 PMCID: PMC11468049 DOI: 10.1186/s12987-024-00580-2] [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: 05/21/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Three common isoforms of the apolipoprotein E (APOE) gene - APOE2, APOE3, and APOE4 - hold varying significance in Alzheimer's Disease (AD) risk. The APOE4 allele is the strongest known genetic risk factor for late-onset Alzheimer's Disease (AD), and its expression has been shown to correlate with increased central nervous system (CNS) amyloid deposition and accelerated neurodegeneration. Conversely, APOE2 is associated with reduced AD risk and lower CNS amyloid burden. Recent clinical data have suggested that increased blood-brain barrier (BBB) leakage is commonly observed among AD patients and APOE4 carriers. However, it remains unclear how different APOE isoforms may impact AD-related pathologies at the BBB. METHODS To explore potential impacts of APOE genotypes on BBB properties and BBB interactions with amyloid beta, we differentiated isogenic human induced pluripotent stem cell (iPSC) lines with different APOE genotypes into both brain microvascular endothelial cell-like cells (BMEC-like cells) and brain pericyte-like cells. We then compared the effect of different APOE isoforms on BBB-related and AD-related phenotypes. Statistical significance was determined via ANOVA with Tukey's post hoc testing as appropriate. RESULTS Isogenic BMEC-like cells with different APOE genotypes had similar trans-endothelial electrical resistance, tight junction integrity and efflux transporter gene expression. However, recombinant APOE4 protein significantly impeded the "brain-to-blood" amyloid beta 1-40 (Aβ40) transport capabilities of BMEC-like cells, suggesting a role in diminished amyloid clearance. Conversely, APOE2 increased amyloid beta 1-42 (Aβ42) transport in the model. Furthermore, we demonstrated that APOE-mediated amyloid transport by BMEC-like cells is dependent on LRP1 and p-glycoprotein pathways, mirroring in vivo findings. Pericyte-like cells exhibited similar APOE secretion levels across genotypes, yet APOE4 pericyte-like cells showed heightened extracellular amyloid deposition, while APOE2 pericyte-like cells displayed the least amyloid deposition, an observation in line with vascular pathologies in AD patients. CONCLUSIONS While APOE genotype did not directly impact general BMEC or pericyte properties, APOE4 exacerbated amyloid clearance and deposition at the model BBB. Conversely, APOE2 demonstrated a potentially protective role by increasing amyloid transport and decreasing deposition. Our findings highlight that iPSC-derived BBB models can potentially capture amyloid pathologies at the BBB, motivating further development of such in vitro models in AD modeling and drug development.
Collapse
Affiliation(s)
- Yunfeng Ding
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
4
|
Liu L, Li J, Zhao C, Qu X, Liu X, Wang D, Wei J. The cellular expression patterns of gdnfa and gdnfb in the gonads of Nile tilapia and their differential response to retinoic acid. Theriogenology 2024; 224:1-8. [PMID: 38714023 DOI: 10.1016/j.theriogenology.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/14/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024]
Abstract
In mammals, glial cell derived neurotrophic factor (GDNF) plays a critical role in the self-renewal and maintenance of spermatogonial stem cells (SSCs) in testis and oogenesis in ovary, whilst retinoic acid (RA), the key factor of meiosis initiation, can downregulate its expression. Unlike mammals, two Gdnf replication genes are widely present in teleost fishes, however, our understanding of them is still poor. In the present study, two paralogous gdnf from Nile tilapia (Oreochromis niloticus), namely as Ongdnfa and Ongdnfb, were characterized, and then their cellular expression profiles in testis and ovary and responsiveness to RA treatment at the tissue and cellular levels were investigated. In phylogenetic tree, the Gdnfa and Gdnfb from teleost fishes were clustered into two different subclasses, respectively, and then clustered with the homologs from cartilaginous fish and tetrapods, suggesting that OnGdnfa and OnGdnfb are orthologous to GDNF and paralogous to each other. Ongdnfa is expressed in Sertoli cells and Leydig cells in testis and oocytes in ovary. The expression pattern of Ongdnfb is similar to Ongdnfa. In the ex vivo testicular organ culture, RA down-regulated the expression of Ongdnfa, whereas up-regulated the expression of Ongdnfb (P < 0.05), suggesting that they have differential responsiveness to RA signaling. RA treatment of the cultured cells derived from adult Nile tilapia testis which have the expression of RA receptors (RAR), Ongdnfa and Ongdnfb further confirmed the above result. Collectively, our study suggests that Ongdnfa and Ongdnfb have non-germline expression patterns in testis and germline expression patterns in ovary; furthermore, they have differential responsiveness to RA signaling, implying that they might have differential biological functions. This study broadens and enriches our understanding of fish GDNF homologs and lays foundation for the study of their biological functions in the future.
Collapse
Affiliation(s)
- Lei Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Jianeng Li
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Changle Zhao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Ximei Qu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China; Chengdu Kangnuoxing Biopharmaceutical Technology Co., Ltd, Chengdu, 610219, China
| | - Xiang Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Deshou Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Jing Wei
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Laboratory of Aquatic Science of Chongqing, School of Life Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
5
|
Deli MA, Porkoláb G, Kincses A, Mészáros M, Szecskó A, Kocsis AE, Vigh JP, Valkai S, Veszelka S, Walter FR, Dér A. Lab-on-a-chip models of the blood-brain barrier: evolution, problems, perspectives. LAB ON A CHIP 2024; 24:1030-1063. [PMID: 38353254 DOI: 10.1039/d3lc00996c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
A great progress has been made in the development and use of lab-on-a-chip devices to model and study the blood-brain barrier (BBB) in the last decade. We present the main types of BBB-on-chip models and their use for the investigation of BBB physiology, drug and nanoparticle transport, toxicology and pathology. The selection of the appropriate cell types to be integrated into BBB-on-chip devices is discussed, as this greatly impacts the physiological relevance and translatability of findings. We identify knowledge gaps, neglected engineering and cell biological aspects and point out problems and contradictions in the literature of BBB-on-chip models, and suggest areas for further studies to progress this highly interdisciplinary field. BBB-on-chip models have an exceptional potential as predictive tools and alternatives of animal experiments in basic and preclinical research. To exploit the full potential of this technique expertise from materials science, bioengineering as well as stem cell and vascular/BBB biology is necessary. There is a need for better integration of these diverse disciplines that can only be achieved by setting clear parameters for characterizing both the chip and the BBB model parts technically and functionally.
Collapse
Affiliation(s)
- Mária A Deli
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Gergő Porkoláb
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - András Kincses
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Mária Mészáros
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Anikó Szecskó
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - Anna E Kocsis
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Judit P Vigh
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
- Doctoral School of Biology, University of Szeged, Hungary
| | - Sándor Valkai
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Szilvia Veszelka
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - Fruzsina R Walter
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| | - András Dér
- HUN-REN Biological Research Centre, Institute of Biophysics, Szeged, Hungary.
| |
Collapse
|
6
|
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
|
7
|
Hudecz D, McCloskey MC, Vergo S, Christensen S, McGrath JL, Nielsen MS. Modelling a Human Blood-Brain Barrier Co-Culture Using an Ultrathin Silicon Nitride Membrane-Based Microfluidic Device. Int J Mol Sci 2023; 24:5624. [PMID: 36982697 PMCID: PMC10058651 DOI: 10.3390/ijms24065624] [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: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Understanding the vesicular trafficking of receptors and receptor ligands in the brain capillary endothelium is essential for the development of the next generations of biologics targeting neurodegenerative diseases. Such complex biological questions are often approached by in vitro models in combination with various techniques. Here, we present the development of a stem cell-based human in vitro blood-brain barrier model composed of induced brain microvascular endothelial cells (iBMECs) on the modular µSiM (a microdevice featuring a silicon nitride membrane) platform. The µSiM was equipped with a 100 nm thick nanoporous silicon nitride membrane with glass-like imaging quality that allowed the use of high-resolution in situ imaging to study the intracellular trafficking. As a proof-of-concept experiment, we investigated the trafficking of two monoclonal antibodies (mAb): an anti-human transferrin receptor mAb (15G11) and an anti-basigin mAb (#52) using the µSiM-iBMEC-human astrocyte model. Our results demonstrated effective endothelial uptake of the selected antibodies; however, no significant transcytosis was observed when the barrier was tight. In contrast, when the iBMECs did not form a confluent barrier on the µSiM, the antibodies accumulated inside both the iBMECs and astrocytes, demonstrating that the cells have an active endocytic and subcellular sorting machinery and that the µSiM itself does not hinder antibody transport. In conclusion, our µSiM-iBMEC-human astrocyte model provides a tight barrier with endothelial-like cells, which can be used for high-resolution in situ imaging and for studying receptor-mediated transport and transcytosis in a physiological barrier.
Collapse
Affiliation(s)
- Diana Hudecz
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Sandra Vergo
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| | - Søren Christensen
- Biotherapeutic Discovery, H. Lundbeck A/S, Valby, 2500 Copenhagen, Denmark
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | | |
Collapse
|
8
|
Linville RM, Sklar MB, Grifno GN, Nerenberg RF, Zhou J, Ye R, DeStefano JG, Guo Z, Jha R, Jamieson JJ, Zhao N, Searson PC. Three-dimensional microenvironment regulates gene expression, function, and tight junction dynamics of iPSC-derived blood-brain barrier microvessels. Fluids Barriers CNS 2022; 19:87. [PMID: 36333694 PMCID: PMC9636829 DOI: 10.1186/s12987-022-00377-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/03/2022] [Indexed: 11/08/2022] Open
Abstract
The blood-brain barrier (BBB) plays a pivotal role in brain health and disease. In the BBB, brain microvascular endothelial cells (BMECs) are connected by tight junctions which regulate paracellular transport, and express specialized transporter systems which regulate transcellular transport. However, existing in vitro models of the BBB display variable accuracy across a wide range of characteristics including gene/protein expression and barrier function. Here, we use an isogenic family of fluorescently-labeled iPSC-derived BMEC-like cells (iBMECs) and brain pericyte-like cells (iPCs) within two-dimensional confluent monolayers (2D) and three-dimensional (3D) tissue-engineered microvessels to explore how 3D microenvironment regulates gene expression and function of the in vitro BBB. We show that 3D microenvironment (shear stress, cell-ECM interactions, and cylindrical geometry) increases BBB phenotype and endothelial identity, and alters angiogenic and cytokine responses in synergy with pericyte co-culture. Tissue-engineered microvessels incorporating junction-labeled iBMECs enable study of the real-time dynamics of tight junctions during homeostasis and in response to physical and chemical perturbations.
Collapse
Affiliation(s)
- Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Matthew B Sklar
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielle N Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Renée F Nerenberg
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Justin Zhou
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Robert Ye
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Jackson G DeStefano
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Ria Jha
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John J Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
Pervaiz I, Zahra FT, Mikelis C, Al-Ahmad AJ. An in vitro model of glucose transporter 1 deficiency syndrome at the blood-brain barrier using induced pluripotent stem cells. J Neurochem 2022; 162:483-500. [PMID: 35943296 DOI: 10.1111/jnc.15684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/08/2022] [Accepted: 08/03/2022] [Indexed: 11/28/2022]
Abstract
Glucose is an important source of energy for the central nervous system. Its uptake at the blood-brain barrier (BBB) is mostly mediated via glucose transporter 1 (GLUT1), a facilitated transporter encoded by the SLC2A1 gene. GLUT1 Deficiency Syndrome (GLUT1DS) is a haploinsufficiency characterized by mutations in the SLC2A1 gene, resulting in impaired glucose uptake at the BBB and clinically characterized by epileptic seizures and movement disorder. A major limitation is an absence of in vitro models of the BBB reproducing the disease. This study aimed to characterize an in vitro model of GLUT1DS using human pluripotent stem cells (iPSCs). Two GLUT1DS clones were generated (GLUT1-iPSC) from their original parental clone iPS(IMR90)-c4 by CRISPR/Cas9 and differentiated into brain microvascular endothelial cells (iBMECs). Cells were characterized in terms of SLC2A1 expression, changes in the barrier function, glucose uptake and metabolism, and angiogenesis. GLUT1DS iPSCs and iBMECs showed comparable phenotype to their parental control, with exception of reduced GLUT1 expression at the protein level. Although no major disruption in the barrier function was reported in the two clones, a significant reduction in glucose uptake accompanied by an increase in glycolysis and mitochondrial respiration was reported in both GLUT1DS-iBMECs. Finally, impaired angiogenic features were reported in such clones compared to the parental clone. Our study provides the first documented characterization of GLUT1DS-iBMECs generated by CRISPR-Cas9, suggesting that GLUT1 truncation appears detrimental to brain angiogenesis and brain endothelial bioenergetics, but maybe not be detrimental to iBMECs differentiation and barriergenesis. Our future direction is to further characterize the functional outcome of such truncated product, as well as its impact on other cells of the neurovascular unit.
Collapse
Affiliation(s)
- Iqra Pervaiz
- Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Department of Pharmaceutical Sciences, Amarillo, Texas, United States of America
| | - Fatema Tuz Zahra
- Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Department of Pharmaceutical Sciences, Amarillo, Texas, United States of America
| | - Constantinos Mikelis
- Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Department of Pharmaceutical Sciences, Amarillo, Texas, United States of America
| | - Abraham Jacob Al-Ahmad
- Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, Department of Pharmaceutical Sciences, Amarillo, Texas, United States of America
| |
Collapse
|
10
|
Linville RM, Nerenberg RF, Grifno G, Arevalo D, Guo Z, Searson PC. Brain microvascular endothelial cell dysfunction in an isogenic juvenile iPSC model of Huntington's disease. Fluids Barriers CNS 2022; 19:54. [PMID: 35773691 PMCID: PMC9245306 DOI: 10.1186/s12987-022-00347-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disease caused by expansion of cytosine-adenine-guanine (CAG) repeats in the huntingtin gene, which leads to neuronal loss and decline in cognitive and motor function. Increasing evidence suggests that blood-brain barrier (BBB) dysfunction may contribute to progression of the disease. Studies in animal models, in vitro models, and post-mortem tissue find that disease progression is associated with increased microvascular density, altered cerebral blood flow, and loss of paracellular and transcellular barrier function. Here, we report on changes in BBB phenotype due to expansion of CAG repeats using an isogenic pair of induced pluripotent stem cells (iPSCs) differentiated into brain microvascular endothelial-like cells (iBMECs). We show that CAG expansion associated with juvenile HD alters the trajectory of iBMEC differentiation, producing cells with ~ two-fold lower percentage of adherent endothelial cells. CAG expansion is associated with diminished transendothelial electrical resistance and reduced tight junction protein expression, but no significant changes in paracellular permeability. While mutant huntingtin protein (mHTT) aggregates were not observed in HD iBMECs, widespread transcriptional dysregulation was observed in iBMECs compared to iPSCs. In addition, CAG expansion in iBMECs results in distinct responses to pathological and therapeutic perturbations including angiogenic factors, oxidative stress, and osmotic stress. In a tissue-engineered BBB model, iBMECs show subtle changes in phenotype, including differences in cell turnover and immune cell adhesion. Our results further support that CAG expansion in BMECs contributes to BBB dysfunction during HD.
Collapse
Affiliation(s)
- Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Renée F Nerenberg
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielle Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Diego Arevalo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
11
|
Pervaiz I, Al-Ahmad AJ. In Vitro Models of the Human Blood-Brain Barrier Utilising Human Induced Pluripotent Stem Cells: Opportunities and Challenges. Methods Mol Biol 2022; 2492:53-72. [PMID: 35733038 DOI: 10.1007/978-1-0716-2289-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) is a component of the neurovascular unit formed by specialized brain microvascular endothelial cells surrounded by astrocytes end-feet processes, pericytes, and a basement membrane. The BBB plays an important role in the maintenance of brain homeostasis and has seen a growing involvement in the pathophysiology of various neurological diseases. On the other hand, the presence of such a barrier remains an important challenge for drug delivery to treat such illnesses.Since the pioneering work describing the isolation and cultivation of primary brain microvascular cells about 50 years ago until now, the development of an in vitro model of the BBB that is scalable, capable to form tight monolayers, and predictive of drug permeability in vivo remained extremely challenging.The recent description of the use of induced pluripotent stem cells (iPSCs) as a modeling tool for neurological diseases raised momentum into the use of such cells to develop new in vitro models of the BBB. This chapter will provide an exhaustive description of the use of iPSCs as a source of cells for modeling the BBB in vitro, describe the advantages and limitations of such model, as well as describe their prospective use for disease modeling and drug permeability screening platforms.
Collapse
Affiliation(s)
- Iqra Pervaiz
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Abraham J Al-Ahmad
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA.
| |
Collapse
|
12
|
Ndemazie NB, Inkoom A, Morfaw EF, Smith T, Aghimien M, Ebesoh D, Agyare E. Multi-disciplinary Approach for Drug and Gene Delivery Systems to the Brain. AAPS PharmSciTech 2021; 23:11. [PMID: 34862567 PMCID: PMC8817187 DOI: 10.1208/s12249-021-02144-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Drug delivery into the brain has for long been a huge challenge as the blood–brain barrier (BBB) offers great resistance to entry of foreign substances (with drugs inclusive) into the brain. This barrier in healthy individuals is protective to the brain, disallowing noxious substances present in the blood to get to the brain while allowing for the exchange of small molecules into the brain by diffusion. However, BBB is disrupted under certain disease conditions, such as cerebrovascular diseases including acute ischemic stroke and intracerebral hemorrhage, and neurodegenerative disorders including multiple sclerosis (MS), Alzheimer’s disease (AD), Parkinson’s disease (PD), and cancers. This review aims to provide a broad overview of present-day strategies for brain drug delivery, emphasizing novel delivery systems. Hopefully, this review would inspire scientists and researchers in the field of drug delivery across BBB to uncover new techniques and strategies to optimize drug delivery to the brain. Considering the anatomy, physiology, and pathophysiological functioning of the BBB in health and disease conditions, this review is focused on the controversies drawn from conclusions of recently published studies on issues such as the penetrability of nanoparticles into the brain, and whether active targeted drug delivery into the brain could be achieved with the use of nanoparticles. We also extended the review to cover novel non-nanoparticle strategies such as using viral and peptide vectors and other non-invasive techniques to enhance brain uptake of drugs.
Collapse
|
13
|
Neal EH, Katdare KA, Shi Y, Marinelli NA, Hagerla KA, Lippmann ES. Influence of basal media composition on barrier fidelity within human pluripotent stem cell-derived blood-brain barrier models. J Neurochem 2021; 159:980-991. [PMID: 34716922 DOI: 10.1111/jnc.15532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 01/02/2023]
Abstract
It is increasingly recognized that brain microvascular endothelial cells (BMECs), the principal component of the blood-brain barrier (BBB), are highly sensitive to soluble cues from both the bloodstream and the brain. This concept extends in vitro, where the extracellular milieu can also influence BBB properties in cultured cells. However, the extent to which baseline culture conditions can affect BBB properties in vitro remains unclear, which has implications for model variability and reproducibility, as well as downstream assessments of molecular transport and disease phenotypes. Here, we explore this concept by examining BBB properties within human-induced pluripotent stem cell (iPSC)-derived BMEC-like cells cultured under serum-free conditions in DMEM/F12 and Neurobasal media, which have fully defined compositions. We demonstrate notable differences in both passive and active BBB properties as a function of basal media composition. Further, RNA sequencing and phosphoproteome analyses revealed alterations to various signaling pathways in response to basal media differences. Overall, our results demonstrate that baseline culture conditions can have a profound influence on the performance of in vitro BBB models, and these effects should be considered when designing experiments that utilize such models for basic research and preclinical assays.
Collapse
Affiliation(s)
- Emma H Neal
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Ketaki A Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Yajuan Shi
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Nicholas A Marinelli
- Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Kameron A Hagerla
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee, USA.,Chemical and Physical Biology Program, Vanderbilt University, Nashville, Tennessee, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Influence of Substrate Stiffness on Barrier Function in an iPSC-Derived In Vitro Blood-Brain Barrier Model. Cell Mol Bioeng 2021; 15:31-42. [DOI: 10.1007/s12195-021-00706-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022] Open
|
15
|
Nguyen J, Lin YY, Gerecht S. The next generation of endothelial differentiation: Tissue-specific ECs. Cell Stem Cell 2021; 28:1188-1204. [PMID: 34081899 DOI: 10.1016/j.stem.2021.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) sense and respond to fluid flow and regulate immune cell trafficking in all organs. Despite sharing the same mesodermal origin, ECs exhibit heterogeneous tissue-specific characteristics. Human pluripotent stem cells (hPSCs) can potentially be harnessed to capture this heterogeneity and further elucidate endothelium behavior to satisfy the need for increased accuracy and breadth of disease models and therapeutics. Here, we review current strategies for hPSC differentiation to blood vascular ECs and their maturation into continuous, fenestrated, and sinusoidal tissues. We then discuss the contribution of hPSC-derived ECs to recent advances in organoid development and organ-on-chip approaches.
Collapse
Affiliation(s)
- Jane Nguyen
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ying-Yu Lin
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology, Physical Sciences-Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
16
|
Lu TM, Barcia Durán JG, Houghton S, Rafii S, Redmond D, Lis R. Human Induced Pluripotent Stem Cell-Derived Brain Endothelial Cells: Current Controversies. Front Physiol 2021; 12:642812. [PMID: 33868008 PMCID: PMC8044318 DOI: 10.3389/fphys.2021.642812] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Brain microvascular endothelial cells (BMECs) possess unique properties that are crucial for many functions of the blood-brain-barrier (BBB) including maintenance of brain homeostasis and regulation of interactions between the brain and immune system. The generation of a pure population of putative brain microvascular endothelial cells from human pluripotent stem cell sources (iBMECs) has been described to meet the need for reliable and reproducible brain endothelial cells in vitro. Human pluripotent stem cells (hPSCs), embryonic or induced, can be differentiated into large quantities of specialized cells in order to study development and model disease. These hPSC-derived iBMECs display endothelial-like properties, such as tube formation and low-density lipoprotein uptake, high transendothelial electrical resistance (TEER), and barrier-like efflux transporter activities. Over time, the de novo generation of an organotypic endothelial cell from hPSCs has aroused controversies. This perspective article highlights the developments made in the field of hPSC derived brain endothelial cells as well as where experimental data are lacking, and what concerns have emerged since their initial description.
Collapse
Affiliation(s)
- Tyler M Lu
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, United States.,Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, United States
| | - José Gabriel Barcia Durán
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, United States
| | - Sean Houghton
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, United States
| | - Shahin Rafii
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, United States
| | - David Redmond
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, United States
| | - Raphaël Lis
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, United States.,Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
17
|
Lu TM, Houghton S, Magdeldin T, Durán JGB, Minotti AP, Snead A, Sproul A, Nguyen DHT, Xiang J, Fine HA, Rosenwaks Z, Studer L, Rafii S, Agalliu D, Redmond D, Lis R. Pluripotent stem cell-derived epithelium misidentified as brain microvascular endothelium requires ETS factors to acquire vascular fate. Proc Natl Acad Sci U S A 2021; 118:e2016950118. [PMID: 33542154 PMCID: PMC7923590 DOI: 10.1073/pnas.2016950118] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cells derived from pluripotent sources in vitro must resemble those found in vivo as closely as possible at both transcriptional and functional levels in order to be a useful tool for studying diseases and developing therapeutics. Recently, differentiation of human pluripotent stem cells (hPSCs) into brain microvascular endothelial cells (ECs) with blood-brain barrier (BBB)-like properties has been reported. These cells have since been used as a robust in vitro BBB model for drug delivery and mechanistic understanding of neurological diseases. However, the precise cellular identity of these induced brain microvascular endothelial cells (iBMECs) has not been well described. Employing a comprehensive transcriptomic metaanalysis of previously published hPSC-derived cells validated by physiological assays, we demonstrate that iBMECs lack functional attributes of ECs since they are deficient in vascular lineage genes while expressing clusters of genes related to the neuroectodermal epithelial lineage (Epi-iBMEC). Overexpression of key endothelial ETS transcription factors (ETV2, ERG, and FLI1) reprograms Epi-iBMECs into authentic endothelial cells that are congruent with bona fide endothelium at both transcriptomic as well as some functional levels. This approach could eventually be used to develop a robust human BBB model in vitro that resembles the human brain EC in vivo for functional studies and drug discovery.
Collapse
Affiliation(s)
- Tyler M Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Tarig Magdeldin
- Department of Neurology and the Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine-New York Presbyterian Hospital, New York, NY 10065
| | - José Gabriel Barcia Durán
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Andrew P Minotti
- Developmental Biology, the Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
- The Biochemistry, Structural Biology, Cell Biology, Developmental Biology and Molecular Biology Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065
| | - Amanda Snead
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Andrew Sproul
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032
| | - Duc-Huy T Nguyen
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY 10065
| | - Howard A Fine
- Department of Neurology and the Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine-New York Presbyterian Hospital, New York, NY 10065
| | - Zev Rosenwaks
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Lorenz Studer
- The Biochemistry, Structural Biology, Cell Biology, Developmental Biology and Molecular Biology Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065
| | - Dritan Agalliu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032
| | - David Redmond
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065;
| | - Raphaël Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065;
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
18
|
Rapid Development of Optic Disc Edema Secondary to Topical Retinoid Cream. Can J Neurol Sci 2021; 48:882-883. [DOI: 10.1017/cjn.2021.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
19
|
Commentary on human pluripotent stem cell-based blood-brain barrier models. Fluids Barriers CNS 2020; 17:64. [PMID: 33076946 PMCID: PMC7574179 DOI: 10.1186/s12987-020-00222-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/03/2020] [Indexed: 01/08/2023] Open
Abstract
In 2012, we provided the first published evidence that human pluripotent stem cells could be differentiated to cells exhibiting markers and phenotypes characteristic of the blood–brain barrier (BBB). In the ensuing years, the initial protocols have been refined, and the research community has identified both positive and negative attributes of this stem cell-based BBB model system. Here, we give our perspective on the current status of these models and their use in the BBB community, as well as highlight key attributes that would benefit from improvement moving forward.
Collapse
|
20
|
Bicker J, Alves G, Fonseca C, Falcão A, Fortuna A. Repairing blood-CNS barriers: Future therapeutic approaches for neuropsychiatric disorders. Pharmacol Res 2020; 162:105226. [PMID: 33007420 DOI: 10.1016/j.phrs.2020.105226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022]
Abstract
Central nervous system (CNS) drug development faces significant difficulties that translate into high rates of failure and lack of innovation. The pathophysiology of neurological and psychiatric disorders often results in the breakdown of blood-CNS barriers, disturbing the CNS microenvironment and worsening disease progression. Therefore, restoring the integrity of blood-CNS barriers may have a beneficial influence in several CNS disorders and improve treatment outcomes. In this review, pathways that may be modulated to protect blood-CNS barriers from neuroinflammatory and oxidative insults are featured. First, the participation of the brain endothelium and glial cells in disruption processes is discussed. Then, the relevance of regulatory systems is analysed, specifically the hypothalamic-pituitary axis, the renin-angiotensin system, sleep and circadian rhythms, and glutamate neurotransmission. Lastly, compounds of endogenous and exogenous origin that are known to mediate the repair of blood-CNS barriers are presented. We believe that enhancing the protection of blood-CNS barriers is a promising therapeutic strategy to pursue in the future.
Collapse
Affiliation(s)
- Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| | - Gilberto Alves
- CICS-UBI, Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Carla Fonseca
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| |
Collapse
|
21
|
Linville RM, DeStefano JG, Nerenberg RF, Grifno GN, Ye R, Gallagher E, Searson PC. Long-Term Cryopreservation Preserves Blood-Brain Barrier Phenotype of iPSC-Derived Brain Microvascular Endothelial Cells and Three-Dimensional Microvessels. Mol Pharm 2020; 17:3425-3434. [PMID: 32787285 PMCID: PMC9923881 DOI: 10.1021/acs.molpharmaceut.0c00484] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain microvascular endothelial cells derived from induced pluripotent stem cells (dhBMECs) are a scalable and reproducible resource for studies of the human blood-brain barrier, including mechanisms and strategies for drug delivery. Confluent monolayers of dhBMECs recapitulate key in vivo functions including tight junctions to limit paracellular permeability and efflux and nutrient transport to regulate transcellular permeability. Techniques for cryopreservation of dhBMECs have been reported; however, functional validation studies after long-term cryopreservation have not been extensively performed. Here, we characterize dhBMECs after 1 year of cryopreservation using selective purification on extracellular matrix-treated surfaces and ROCK inhibition. One-year cryopreserved dhBMECs maintain functionality of tight junctions, efflux pumps, and nutrient transporters with stable protein localization and gene expression. Cryopreservation is associated with a decrease in the yield of adherent cells and unique responses to cell stress, resulting in altered paracellular permeability of Lucifer yellow. Additionally, cryopreserved dhBMECs reliably form functional three-dimensional microvessels independent of cryopreservation length, with permeabilities lower than non-cryopreserved two-dimensional models. Long-term cryopreservation of dhBMECs offers key advantages including increased scalability, reduced batch-to-batch effects, the ability to conduct well-controlled follow up studies, and support of multisite collaboration from the same cell stock, all while maintaining phenotype for screening pharmaceutical agents.
Collapse
Affiliation(s)
- Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Jackson G. DeStefano
- Institute for Nanobiotechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Reneé F. Nerenberg
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Gabrielle N. Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| | - Robert Ye
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Erin Gallagher
- Institute for Nanobiotechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21218, United States
| |
Collapse
|
22
|
Ko HJ, Hong SW, Verma R, Jung J, Lee M, Kim N, Kim D, Surh CD, Kim KS, Rudra D, Im SH. Dietary Glucose Consumption Promotes RALDH Activity in Small Intestinal CD103 +CD11b + Dendritic Cells. Front Immunol 2020; 11:1897. [PMID: 32849649 PMCID: PMC7433714 DOI: 10.3389/fimmu.2020.01897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022] Open
Abstract
Retinal dehydrogenase (RALDH) enzymatic activities catalyze the conversion of vitamin A to its metabolite Retinoic acid (RA) in intestinal dendritic cells (DCs) and promote immunological tolerance. However, precise understanding of the exogenous factors that act as initial trigger of RALDH activity in these cells is still evolving. By using germ-free (GF) mice raised on an antigen free (AF) elemental diet, we find that certain components in diet are critically required to establish optimal RALDH expression and activity, most prominently in small intestinal CD103+CD11b+ DCs (siLP-DCs) right from the beginning of their lives. Surprisingly, systematic screens using modified diets devoid of individual dietary components indicate that proteins, starch and minerals are dispensable for this activity. On the other hand, in depth comparison between subtle differences in dietary composition among different dietary regimes reveal that adequate glucose concentration in diet is a critical determinant for establishing RALDH activity specifically in siLP-DCs. Consequently, pre-treatment of siLP-DCs, and not mesenteric lymph node derived MLNDCs with glucose, results in significant enhancement in the in vitro generation of induced Regulatory T (iTreg) cells. Our findings reveal previously underappreciated role of dietary glucose concentration in establishing regulatory properties in intestinal DCs, thereby extending a potential therapeutic module against intestinal inflammation.
Collapse
Affiliation(s)
- Hyun-Ja Ko
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang-si, South Korea
| | - Sung-Wook Hong
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang-si, South Korea
| | - Ravi Verma
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea.,ImmunoBiome Inc., Pohang-si, South Korea
| | - Jisun Jung
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Minji Lee
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Nahyun Kim
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Daeun Kim
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Charles D Surh
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang-si, South Korea.,Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Kwang Soon Kim
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Dipayan Rudra
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea
| | - Sin-Hyeog Im
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology, Pohang-si, South Korea.,ImmunoBiome Inc., Pohang-si, South Korea
| |
Collapse
|
23
|
Recent advances in human iPSC-derived models of the blood-brain barrier. Fluids Barriers CNS 2020; 17:30. [PMID: 32321511 PMCID: PMC7178976 DOI: 10.1186/s12987-020-00191-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/07/2020] [Indexed: 01/07/2023] Open
Abstract
The blood–brain barrier (BBB) is a critical component of the central nervous system that protects neurons and other cells of the brain parenchyma from potentially harmful substances found in peripheral circulation. Gaining a thorough understanding of the development and function of the human BBB has been hindered by a lack of relevant models given significant species differences and limited access to in vivo tissue. However, advances in induced pluripotent stem cell (iPSC) and organ-chip technologies now allow us to improve our knowledge of the human BBB in both health and disease. This review focuses on the recent progress in modeling the BBB in vitro using human iPSCs.
Collapse
|
24
|
A Simplified, Fully Defined Differentiation Scheme for Producing Blood-Brain Barrier Endothelial Cells from Human iPSCs. Stem Cell Reports 2020; 12:1380-1388. [PMID: 31189096 PMCID: PMC6565873 DOI: 10.1016/j.stemcr.2019.05.008] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived developmental lineages are key tools for in vitro mechanistic interrogations, drug discovery, and disease modeling. iPSCs have previously been differentiated to endothelial cells with blood-brain barrier (BBB) properties, as defined by high transendothelial electrical resistance (TEER), low passive permeability, and active transporter functions. Typical protocols use undefined components, which impart unacceptable variability on the differentiation process. We demonstrate that replacement of serum with fully defined components, from common medium supplements to a simple mixture of insulin, transferrin, and selenium, yields BBB endothelium with TEER in the range of 2,000-8,000 Ω × cm2 across multiple iPSC lines, with appropriate marker expression and active transporters. The use of a fully defined medium vastly improves the consistency of differentiation, and co-culture of BBB endothelium with iPSC-derived astrocytes produces a robust in vitro neurovascular model. This defined differentiation scheme should broadly enable the use of human BBB endothelium for diverse applications.
Collapse
|
25
|
Yang CD, Cheng ML, Liu W, Zeng DH. Association of serum retinoic acid with depression in patients with acute ischemic stroke. Aging (Albany NY) 2020; 12:2647-2658. [PMID: 32040942 PMCID: PMC7041768 DOI: 10.18632/aging.102767] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/12/2020] [Indexed: 12/17/2022]
Abstract
Retinoic acid (RA), produced by the metabolism of vitamin A, makes effects on depression and stroke. This study was aimed to evaluate the relationship between RA levels in serum and post-stroke depression (PSD). A single-center (Chengdu, China) prospective cohort study was conducted on patients with acute ischemic stroke. The RA serum level was measured at admission. The PSD was assessed in the 3-month follow-up. The RA-PSD relationship was evaluated with conditional logistic regression. In total, 239 ischemic stroke cases and 100 healthy controls were included. The median RA serum level in patients with ischemic stroke was 2.45 ng/ml (interquartile range [IQR], 0.72-4.33), lower(P<0.001) than 3.89 ng/ml of those in control cases ([IQR]: 2.62-5.39). The crude and adjusted odds ratios [OR] (and 95% confidence intervals [CI]) of PSD associated with an IQR increase for RA were 0.54 (0.44, 0.67) and 0.66 (0.52, 0.79), respectively. Higher ORs of PSD associated with reduced RA levels (
Collapse
Affiliation(s)
- Cai-Di Yang
- Department of Neurology, Eastern Hospital, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610101, China
| | - Ming-Li Cheng
- Department of Neurology, People's Hospital of Jianyang, Jianyang 641400, China
| | - Wen Liu
- The Clinical Laboratory Department, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Ding-Hua Zeng
- Department of Neurology, Eastern Hospital, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610101, China
| |
Collapse
|
26
|
Abstract
Knowledge about the transport of active compounds across the blood-brain barrier is of essential importance for drug development. Systemically applied drugs for the central nervous system (CNS) must be able to cross the blood-brain barrier in order to reach their target sites, whereas drugs that are supposed to act in the periphery should not permeate the blood-brain barrier so that they do not trigger any adverse central adverse effects. A number of approaches have been pursued, and manifold in silico, in vitro, and in vivo animal models were developed in order to be able to make a better prediction for humans about the possible penetration of active substances into the CNS. In this particular case, however, in vitro models play a special role, since the data basis for in silico models is usually in need of improvement, and the predictive power of in vivo animal models has to be checked for possible species differences. The blood-brain barrier is a dynamic, highly selective barrier formed by brain capillary endothelial cells. One of its main tasks is the maintenance of homeostasis in the CNS. The function of the barrier is regulated by cells of the microenvironment and the shear stress mediated by the blood flow, which makes the model development most complex. In general, one could follow the credo "as easy as possible, as complex as necessary" for the usage of in vitro BBB models for drug development. In addition to the description of the classical cell culture models (transwell, hollow fiber) and guidance how to apply them, the latest developments (spheroids, microfluidic models) will be introduced in this chapter, as it is attempted to get more in vivo-like and to be applicable for high-throughput usage with these models. Moreover, details about the development of models based on stem cells derived from different sources with a special focus on human induced pluripotent stem cells are presented.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Competence Unit Molecular Diagnostics, Center Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria.
| |
Collapse
|
27
|
Al-Ahmad AJ, Patel R, Palecek SP, Shusta EV. Hyaluronan impairs the barrier integrity of brain microvascular endothelial cells through a CD44-dependent pathway. J Cereb Blood Flow Metab 2019; 39:1759-1775. [PMID: 29589805 PMCID: PMC6727144 DOI: 10.1177/0271678x18767748] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hyaluronan (HA) constitutes the most abundant extracellular matrix component during brain development, only to become a minor component rapidly after birth and in adulthood to remain in specified regions. HA signaling has been associated with several neurological disorders, yet the impact of HA signaling at the blood-brain barrier (BBB) function remains undocumented. In this study, we investigated the impact of HA on BBB properties using human-induced pluripotent stem cell (iPSC) -derived and primary human and rat BMECs. The impact of HA signaling on developmental and mature BMECs was assessed by measuring changes in TEER, permeability, BMECs markers (GLUT1, tight junction proteins, P-gp) expression and localization, CD44 expression and hyaluronan levels. In general, HA treatment decreased barrier function and reduced P-gp activity with effects being more prominent upon treatment with oligomeric forms of HA (oHA). Such effects were exacerbated when applied during BMEC differentiation phase (considered as developmental BBB). We noted a hyaluronidase activity as well as an increase in CD44 expression during prolonged oxygen-glucose deprivation stress. Inhibition of HA signaling by antibody blockade of CD44 abrogated the detrimental effects of HA treatment. These results suggest the importance of HA signaling through CD44 on BBB properties.
Collapse
Affiliation(s)
- Abraham J Al-Ahmad
- 1 Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA.,2 Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Ronak Patel
- 2 Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Sean P Palecek
- 1 Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- 1 Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
28
|
Canfield SG, Stebbins MJ, Faubion MG, Gastfriend BD, Palecek SP, Shusta EV. An isogenic neurovascular unit model comprised of human induced pluripotent stem cell-derived brain microvascular endothelial cells, pericytes, astrocytes, and neurons. Fluids Barriers CNS 2019; 16:25. [PMID: 31387594 PMCID: PMC6685239 DOI: 10.1186/s12987-019-0145-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/09/2019] [Indexed: 11/28/2022] Open
Abstract
Background Brain microvascular endothelial cells (BMECs) astrocytes, neurons, and pericytes form the neurovascular unit (NVU). Interactions with NVU cells endow BMECs with extremely tight barriers via the expression of tight junction proteins, a host of active efflux and nutrient transporters, and reduced transcellular transport. To recreate the BMEC-enhancing functions of NVU cells, we combined BMECs, astrocytes, neurons, and brain pericyte-like cells. Methods BMECs, neurons, astrocytes, and brain like pericytes were differentiated from human induced pluripotent stem cells (iPSCs) and placed in a Transwell-type NVU model. BMECs were placed in co-culture with neurons, astrocytes, and/or pericytes alone or in varying combinations and critical barrier properties were monitored. Results Co-culture with pericytes followed by a mixture of neurons and astrocytes (1:3) induced the greatest barrier tightening in BMECs, supported by a significant increase in junctional localization of occludin. BMECs also expressed active P-glycoprotein (PGP) efflux transporters under baseline BMEC monoculture conditions and continued to express baseline active PGP efflux transporters regardless of co-culture conditions. Finally, brain-like pericyte co-culture significantly reduced the rate of non-specific transcytosis across BMECs. Conclusions Importantly, each cell type in the NVU model was differentiated from the same donor iPSC source, yielding an isogenic model that could prove enabling for enhanced personalized modeling of the NVU in human health and disease.
Collapse
Affiliation(s)
- Scott G Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA. .,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 620 Chestnut Street, Terre Haute, IN, 47809, USA.
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Madeline G Faubion
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Benjamin D Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| |
Collapse
|
29
|
Studying Heterotypic Cell⁻Cell Interactions in the Human Brain Using Pluripotent Stem Cell Models for Neurodegeneration. Cells 2019; 8:cells8040299. [PMID: 30939814 PMCID: PMC6523455 DOI: 10.3390/cells8040299] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 02/08/2023] Open
Abstract
Human cerebral organoids derived from induced pluripotent stem cells (iPSCs) provide novel tools for recapitulating the cytoarchitecture of the human brain and for studying biological mechanisms of neurological disorders. However, the heterotypic interactions of neurovascular units, composed of neurons, pericytes (i.e., the tissue resident mesenchymal stromal cells), astrocytes, and brain microvascular endothelial cells, in brain-like tissues are less investigated. In addition, most cortical organoids lack a microglia component, the resident immune cells in the brain. Impairment of the blood-brain barrier caused by improper crosstalk between neural cells and vascular cells is associated with many neurodegenerative disorders. Mesenchymal stem cells (MSCs), with a phenotype overlapping with pericytes, have promotion effects on neurogenesis and angiogenesis, which are mainly attributed to secreted growth factors and extracellular matrices. As the innate macrophages of the central nervous system, microglia regulate neuronal activities and promote neuronal differentiation by secreting neurotrophic factors and pro-/anti-inflammatory molecules. Neuronal-microglia interactions mediated by chemokines signaling can be modulated in vitro for recapitulating microglial activities during neurodegenerative disease progression. In this review, we discussed the cellular interactions and the physiological roles of neural cells with other cell types including endothelial cells and microglia based on iPSC models. The therapeutic roles of MSCs in treating neural degeneration and pathological roles of microglia in neurodegenerative disease progression were also discussed.
Collapse
|
30
|
Pawlikowski B, Wragge J, Siegenthaler JA. Retinoic acid signaling in vascular development. Genesis 2019; 57:e23287. [PMID: 30801891 DOI: 10.1002/dvg.23287] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
Formation of the vasculature is an essential developmental process, delivering oxygen and nutrients to support cellular processes needed for tissue growth and maturation. Retinoic acid (RA) and its downstream signaling pathway is vital for normal pre- and post-natal development, playing key roles in the specification and formation of many organs and tissues. Here, we review the role of RA in blood and lymph vascular development, beginning with embryonic yolk sac vasculogenesis and remodeling and discussing RA's organ-specific roles in angiogenesis and vessel maturation. In particular, we highlight the multi-faceted role of RA signaling in CNS vascular development and acquisition of blood-brain barrier properties.
Collapse
Affiliation(s)
- Brad Pawlikowski
- Department of Molecular, Cell and Developmental Biology, University of Colorado-Boulder, Boulder, Colorado
| | - Jacob Wragge
- Department of Pediatrics-Section of Developmental Biology, University of Colorado, School of Medicine-Anschutz Medical Campus, Aurora, Colorado
| | - Julie A Siegenthaler
- Department of Pediatrics-Section of Developmental Biology, University of Colorado, School of Medicine-Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
31
|
Stebbins MJ, Gastfriend BD, Canfield SG, Lee MS, Richards D, Faubion MG, Li WJ, Daneman R, Palecek SP, Shusta EV. Human pluripotent stem cell-derived brain pericyte-like cells induce blood-brain barrier properties. SCIENCE ADVANCES 2019; 5:eaau7375. [PMID: 30891496 PMCID: PMC6415958 DOI: 10.1126/sciadv.aau7375] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/29/2019] [Indexed: 05/18/2023]
Abstract
Brain pericytes play important roles in the formation and maintenance of the neurovascular unit (NVU), and their dysfunction has been implicated in central nervous system disorders. While human pluripotent stem cells (hPSCs) have been used to model other NVU cell types, including brain microvascular endothelial cells (BMECs), astrocytes, and neurons, hPSC-derived brain pericyte-like cells have not been integrated into these models. In this study, we generated neural crest stem cells (NCSCs), the embryonic precursor to forebrain pericytes, from hPSCs and subsequently differentiated NCSCs to brain pericyte-like cells. These cells closely resembled primary human brain pericytes and self-assembled with endothelial cells. The brain pericyte-like cells induced blood-brain barrier properties in BMECs, including barrier enhancement and reduced transcytosis. Last, brain pericyte-like cells were incorporated with iPSC-derived BMECs, astrocytes, and neurons to form an isogenic human model that should prove useful for the study of the NVU.
Collapse
Affiliation(s)
- Matthew J. Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Benjamin D. Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Scott G. Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Ming-Song Lee
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Drew Richards
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Madeline G. Faubion
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Richard Daneman
- Departments of Neuroscience and Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Eric V. Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
32
|
Keep RF, Jones HC, Drewes LR. The year in review: progress in brain barriers and brain fluid research in 2018. Fluids Barriers CNS 2019; 16:4. [PMID: 30717760 PMCID: PMC6362595 DOI: 10.1186/s12987-019-0124-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 12/17/2022] Open
Abstract
This editorial focuses on the progress made in brain barrier and brain fluid research in 2018. It highlights some recent advances in knowledge and techniques, as well as prevalent themes and controversies. Areas covered include: modeling, the brain endothelium, the neurovascular unit, the blood–CSF barrier and CSF, drug delivery, fluid movement within the brain, the impact of disease states, and heterogeneity.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| | - Hazel C Jones
- Gagle Brook House, Chesterton, Bicester, OX26 1UF, UK
| | - Lester R Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN, 55812, USA
| |
Collapse
|
33
|
DeStefano JG, Jamieson JJ, Linville RM, Searson PC. Benchmarking in vitro tissue-engineered blood-brain barrier models. Fluids Barriers CNS 2018; 15:32. [PMID: 30514389 PMCID: PMC6280508 DOI: 10.1186/s12987-018-0117-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022] Open
Abstract
The blood–brain barrier (BBB) plays a key role in regulating transport into and out of the brain. With increasing interest in the role of the BBB in health and disease, there have been significant advances in the development of in vitro models. The value of these models to the research community is critically dependent on recapitulating characteristics of the BBB in humans or animal models. However, benchmarking in vitro models is surprisingly difficult since much of our knowledge of the structure and function of the BBB comes from in vitro studies. Here we describe a set of parameters that we consider a starting point for benchmarking and validation. These parameters are associated with structure (ultrastructure, wall shear stress, geometry), microenvironment (basement membrane and extracellular matrix), barrier function (transendothelial electrical resistance, permeability, efflux transport), cell function (expression of BBB markers, turnover), and co-culture with other cell types (astrocytes and pericytes). In suggesting benchmarks, we rely primarily on imaging or direct measurements in humans and animal models.
Collapse
Affiliation(s)
- Jackson G DeStefano
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John J Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA. .,120 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
34
|
Impact of Retinoic Acid on Immune Cells and Inflammatory Diseases. Mediators Inflamm 2018; 2018:3067126. [PMID: 30158832 PMCID: PMC6109577 DOI: 10.1155/2018/3067126] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/16/2018] [Accepted: 06/28/2018] [Indexed: 12/14/2022] Open
Abstract
Vitamin A metabolite retinoic acid (RA) plays important roles in cell growth, differentiation, organogenesis, and reproduction and a key role in mucosal immune responses. RA promotes dendritic cells to express CD103 and to produce RA, enhances the differentiation of Foxp3+ inducible regulatory T cells, and induces gut-homing specificity in T cells. Although vitamin A is crucial for maintaining homeostasis at the intestinal barrier and equilibrating immunity and tolerance, including gut dysbiosis, retinoids perform a wide variety of functions in many settings, such as the central nervous system, skin aging, allergic airway diseases, cancer prevention and therapy, and metabolic diseases. The mechanism of RA is interesting to explore as both a mucosal adjuvant and a combination therapy with other effective agents. Here, we review the effect of RA on innate and adaptive immunity with a special emphasis on inflammatory status.
Collapse
|
35
|
Cabral JMS, Palecek SP. Regenerative Medicine Manufacturing. Biotechnol J 2018; 13. [PMID: 29392889 DOI: 10.1002/biot.201800026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Indexed: 11/06/2022]
|