1
|
Vollmuth N, Bridgers BE, Armstrong ML, Wood JF, Gildea AR, Espinal ER, Hooven TA, Barbieri G, Westermann AJ, Sauerwein T, Foerstner KU, Schubert-Unkmeir A, Kim BJ. Group B Streptococcus transcriptome when interacting with brain endothelial cells. J Bacteriol 2024; 206:e0008724. [PMID: 38771039 PMCID: PMC11332166 DOI: 10.1128/jb.00087-24] [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: 03/01/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Bacterial meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when bacteria are able to cross the blood-brain barrier (BBB) or the meningeal-cerebrospinal fluid barrier (mBCSFB). The BBB and mBCSFB comprise highly specialized brain endothelial cells (BECs) that typically restrict pathogen entry. Group B Streptococcus (GBS or Streptococcus agalactiae) is the leading cause of neonatal meningitis. Until recently, identification of GBS virulence factors has relied on genetic screening approaches. Instead, we here conducted RNA-seq analysis on GBS when interacting with induced pluripotent stem cell-derived BECs (iBECs) to pinpoint virulence-associated genes. Of the 2,068 annotated protein-coding genes of GBS, 430 transcripts displayed significant changes in expression after interacting with BECs. Notably, we found that the majority of differentially expressed GBS transcripts were downregulated (360 genes) during infection of iBECs. Interestingly, codY, encoding a pleiotropic transcriptional repressor in low-G + C Gram-positive bacteria, was identified as being highly downregulated. We conducted qPCR to confirm the codY downregulation observed via RNA-seq during the GBS-iBEC interaction and obtained codY mutants in three different GBS background parental strains. As anticipated from the RNA-seq results, the [Formula: see text]codY strains were more adherent and invasive in two in vitro BEC models. Together, this demonstrates the utility of RNA-seq during the BEC interaction to identify GBS virulence modulators. IMPORTANCE Group B Streptococcus (GBS) meningitis remains the leading cause of neonatal meningitis. Research work has identified surface factors and two-component systems that contribute to GBS disruption of the blood-brain barrier (BBB). These discoveries often relied on genetic screening approaches. Here, we provide transcriptomic data describing how GBS changes its transcriptome when interacting with brain endothelial cells. Additionally, we have phenotypically validated these data by obtaining mutants of a select regulator that is highly down-regulated during infection and testing on our BBB model. This work provides the research field with a validated data set that can provide an insight into potential pathways that GBS requires to interact with the BBB and open the door to new discoveries.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Bailey E. Bridgers
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Madelyn L. Armstrong
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Jacob F. Wood
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Abigail R. Gildea
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Eric R. Espinal
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Thomas A. Hooven
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Richard King Mellon Institute for Pediatric Research, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Alexander J. Westermann
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Wuerzburg, Germany
| | - Till Sauerwein
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- ZB MED, Information Centre for Life Sciences, Cologne, Germany
| | - Konrad U. Foerstner
- Institute of Molecular Infection Biology (IMIB), University of Wuerzburg, Wuerzburg, Germany
- ZB MED, Information Centre for Life Sciences, Cologne, Germany
- TH Koeln, University of Applied Sciences, Cologne, Germany
| | | | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham Heesink School of Medicine, Birmingham, Alabama, USA
- University of Alabama Center of Convergent Biosciences and Medicine, Tuscaloosa, Alabama, USA
- University of Alabama Life Research, Tuscaloosa, Alabama, USA
| |
Collapse
|
2
|
Joof F, Hu R, Saidi A, Seydel KB, Cohee LM, Zheng Y, Smith JD. Plasma from older children in Malawi inhibits Plasmodium falciparum binding in 3D brain microvessels. J Infect Dis 2024:jiae315. [PMID: 38875153 DOI: 10.1093/infdis/jiae315] [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: 03/06/2024] [Revised: 05/02/2024] [Accepted: 06/12/2024] [Indexed: 06/16/2024] Open
Abstract
A hallmark of cerebral malaria is sequestration of Plasmodium falciparum-infected erythrocytes (IEs) in the brain microcirculation. Antibodies contribute to malaria immunity, but it remains unclear whether functional antibodies targeting parasite-expressed ligand can block cytoadhesion in the brain. Here, we screened the plasma of older children and young adults in Malawi to characterize the antibody response against the P. falciparum-IE surface and used a bioengineered 3D human brain microvessel model incorporating variable flow dynamics to measure adhesion blocking responses. We found a strong correlation between surface antibody reactivity by flow cytometry and reduced P. falciparum-IE binding in 3D microvessels. Moreover, there was a threshold of surface antibody reactivity necessary to achieve robust inhibitory activity. Our findings provide evidence of the acquisition of adhesion blocking antibodies against cerebral binding variants in people exposed to stable P. falciparum transmission and suggest the quality of the inhibitory response can be influenced by flow dynamics.
Collapse
Affiliation(s)
- Fatou Joof
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA USA
| | - Ruoqian Hu
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Alex Saidi
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Karl B Seydel
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Lauren M Cohee
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ying Zheng
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Joseph D Smith
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA USA
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
3
|
Vollmuth N, Sin J, Kim BJ. Host-microbe interactions at the blood-brain barrier through the lens of induced pluripotent stem cell-derived brain-like endothelial cells. mBio 2024; 15:e0286223. [PMID: 38193670 PMCID: PMC10865987 DOI: 10.1128/mbio.02862-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Abstract
Microbe-induced meningoencephalitis/meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when pathogens are able to cross the blood-brain barrier (BBB) and gain access to the CNS. The BBB consists of highly specialized brain endothelial cells that exhibit specific properties to allow tight regulation of CNS homeostasis and prevent pathogen crossing. However, during meningoencephalitis/meningitis, the BBB fails to protect the CNS. Modeling the BBB remains a challenge due to the specialized characteristics of these cells. In this review, we cover the induced pluripotent stem cell-derived, brain-like endothelial cell model during host-pathogen interaction, highlighting the strengths and recent work on various pathogens known to interact with the BBB. As stem cell technologies are becoming more prominent, the stem cell-derived, brain-like endothelial cell model has been able to reveal new insights in vitro, which remain challenging with other in vitro cell-based models consisting of primary human brain endothelial cells and immortalized human brain endothelial cell lines.
Collapse
Affiliation(s)
- Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, USA
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, Alabama, USA
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, USA
| |
Collapse
|
4
|
Brandl S, Reindl M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int J Mol Sci 2023; 24:12699. [PMID: 37628879 PMCID: PMC10454051 DOI: 10.3390/ijms241612699] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The blood-brain barrier, which is formed by tightly interconnected microvascular endothelial cells, separates the brain from the peripheral circulation. Together with other central nervous system-resident cell types, including pericytes and astrocytes, the blood-brain barrier forms the neurovascular unit. Upon neuroinflammation, this barrier becomes leaky, allowing molecules and cells to enter the brain and to potentially harm the tissue of the central nervous system. Despite the significance of animal models in research, they may not always adequately reflect human pathophysiology. Therefore, human models are needed. This review will provide an overview of the blood-brain barrier in terms of both health and disease. It will describe all key elements of the in vitro models and will explore how different compositions can be utilized to effectively model a variety of neuroinflammatory conditions. Furthermore, it will explore the existing types of models that are used in basic research to study the respective pathologies thus far.
Collapse
Affiliation(s)
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
5
|
Mamana J, Humber GM, Espinal ER, Seo S, Vollmuth N, Sin J, Kim BJ. Coxsackievirus B3 infects and disrupts human induced-pluripotent stem cell derived brain-like endothelial cells. Front Cell Infect Microbiol 2023; 13:1171275. [PMID: 37139492 PMCID: PMC10149843 DOI: 10.3389/fcimb.2023.1171275] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Coxsackievirus B3 (CVB3) is a significant human pathogen that is commonly found worldwide. CVB3 among other enteroviruses, are the leading causes of aseptic meningo-encephalitis which can be fatal especially in young children. How the virus gains access to the brain is poorly-understood, and the host-virus interactions that occur at the blood-brain barrier (BBB) is even less-characterized. The BBB is a highly specialized biological barrier consisting primarily of brain endothelial cells which possess unique barrier properties and facilitate the passage of nutrients into the brain while restricting access to toxins and pathogens including viruses. To determine the effects of CVB3 infection on the BBB, we utilized a model of human induced-pluripotent stem cell-derived brain-like endothelial cells (iBECs) to ascertain if CVB3 infection may alter barrier cell function and overall survival. In this study, we determined that these iBECs indeed are susceptible to CVB3 infection and release high titers of extracellular virus. We also determined that infected iBECs maintain high transendothelial electrical resistance (TEER) during early infection despite possessing high viral load. TEER progressively declines at later stages of infection. Interestingly, despite the high viral burden and TEER disruptions at later timepoints, infected iBEC monolayers remain intact, indicating a low degree of late-stage virally-mediated cell death, which may contribute to prolonged viral shedding. We had previously reported that CVB3 infections rely on the activation of transient receptor vanilloid potential 1 (TRPV1) and found that inhibiting TRPV1 activity with SB-366791 significantly limited CVB3 infection of HeLa cervical cancer cells. Similarly in this study, we observed that treating iBECs with SB-366791 significantly reduced CVB3 infection, which suggests that not only can this drug potentially limit viral entry into the brain, but also demonstrates that this infection model could be a valuable platform for testing antiviral treatments of neurotropic viruses. In all, our findings elucidate the unique effects of CVB3 infection on the BBB and shed light on potential mechanisms by which the virus can initiate infections in the brain.
Collapse
Affiliation(s)
- Julia Mamana
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Gabrielle M. Humber
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Eric R. Espinal
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Soojung Seo
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- *Correspondence: Jon Sin, ; Brandon J. Kim,
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, AL, United States
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, United States
- *Correspondence: Jon Sin, ; Brandon J. Kim,
| |
Collapse
|
6
|
Endres LM, Jungblut M, Divyapicigil M, Sauer M, Stigloher C, Christodoulides M, Kim BJ, Schubert-Unkmeir A. Development of a multicellular in vitro model of the meningeal blood-CSF barrier to study Neisseria meningitidis infection. Fluids Barriers CNS 2022; 19:81. [PMID: 36289516 PMCID: PMC9597984 DOI: 10.1186/s12987-022-00379-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/06/2022] [Indexed: 12/01/2022] Open
Abstract
Background Bacterial meningitis is a life-threatening disease that occurs when pathogens such as Neisseria meningitidis cross the meningeal blood cerebrospinal fluid barrier (mBCSFB) and infect the meninges. Due to the human-specific nature of N. meningitidis, previous research investigating this complex host–pathogen interaction has mostly been done in vitro using immortalized brain endothelial cells (BECs) alone, which often do not retain relevant barrier properties in culture. Here, we developed physiologically relevant mBCSFB models using BECs in co-culture with leptomeningeal cells (LMCs) to examine N. meningitidis interaction. Methods We used BEC-like cells derived from induced pluripotent stem cells (iBECs) or hCMEC/D3 cells in co-culture with LMCs derived from tumor biopsies. We employed TEM and structured illumination microscopy to characterize the models as well as bacterial interaction. We measured TEER and sodium fluorescein (NaF) permeability to determine barrier tightness and integrity. We then analyzed bacterial adherence and penetration of the cell barrier and examined changes in host gene expression of tight junctions as well as chemokines and cytokines in response to infection. Results Both cell types remained distinct in co-culture and iBECs showed characteristic expression of BEC markers including tight junction proteins and endothelial markers. iBEC barrier function as determined by TEER and NaF permeability was improved by LMC co-culture and remained stable for seven days. BEC response to N. meningitidis infection was not affected by LMC co-culture. We detected considerable amounts of BEC-adherent meningococci and a relatively small number of intracellular bacteria. Interestingly, we discovered bacteria traversing the BEC-LMC barrier within the first 24 h post-infection, when barrier integrity was still high, suggesting a transcellular route for N. meningitidis into the CNS. Finally, we observed deterioration of barrier properties including loss of TEER and reduced expression of cell-junction components at late time points of infection. Conclusions Here, we report, for the first time, on co-culture of human iPSC derived BECs or hCMEC/D3 with meningioma derived LMCs and find that LMC co-culture improves barrier properties of iBECs. These novel models allow for a better understanding of N. meningitidis interaction at the mBCSFB in a physiologically relevant setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00379-z.
Collapse
Affiliation(s)
- Leo M. Endres
- grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| | - Marvin Jungblut
- grid.8379.50000 0001 1958 8658Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Mustafa Divyapicigil
- grid.411015.00000 0001 0727 7545Department of Biological Sciences, University of Alabama, Tuscaloosa, AL USA ,grid.265892.20000000106344187Department of Microbiology Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.411015.00000 0001 0727 7545Center for Convergent Biosciences & Medicine, University of Alabama, Tuscaloosa, AL USA ,grid.411015.00000 0001 0727 7545Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL USA
| | - Markus Sauer
- grid.8379.50000 0001 1958 8658Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Christian Stigloher
- grid.8379.50000 0001 1958 8658Imaging Core Facility, Biocenter, University of Würzburg, Würzburg, Germany
| | - Myron Christodoulides
- grid.5491.90000 0004 1936 9297Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Brandon J. Kim
- grid.411015.00000 0001 0727 7545Department of Biological Sciences, University of Alabama, Tuscaloosa, AL USA ,grid.265892.20000000106344187Department of Microbiology Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA ,grid.411015.00000 0001 0727 7545Center for Convergent Biosciences & Medicine, University of Alabama, Tuscaloosa, AL USA ,grid.411015.00000 0001 0727 7545Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL USA
| | - Alexandra Schubert-Unkmeir
- grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Strasse 2, 97080 Würzburg, Germany
| |
Collapse
|
7
|
Fc-MBL-modified Fe 3O 4 magnetic bead enrichment and fixation in Gram stain for rapid detection of low-concentration bacteria. Mikrochim Acta 2022; 189:169. [PMID: 35364796 DOI: 10.1007/s00604-022-05277-7] [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: 12/25/2021] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Functional bacterial enrichment magnetic beads (Fe3O4@SiO2@Fc-MBL) and Gram staining were combined for the fast diagnosis of infecting bacteria in meningitis. Fe3O4@SiO2@Fc-MBL has excellent microbial binding ability and can be used for bacterial enrichment from cerebrospinal fluid (CSF). The enriched bacteria are recognized by Gram stain at very low concentrations (10 CFU·mL-1). The feasibility of this method was verified by five common bacteria in meningitis infection (Gram-positive: Staphylococcus epidermidis, Staphylococcus haemolyticus, and Staphylococcus capitis; Gram-negative: Klebsiella pneumoniae and Escherichia coli). The extraction efficiency of Fc-MBL-modified Fe3O4 magnetic beads was approximately 90% in artificial CSF for the selected bacteria, with the exception of E. coli (~ 60%). The bacteria were successfully recognized by Gram staining and microscopic observation. Fe3O4@SiO2@Fc-MBL acts by capturing and fixing the bacteria in a magnetic field throughout the experiment. Compared with traditional CSF Gram staining, this new method avoids interference by inflammatory cells and red blood cells during microscopic examination. Furthermore, the sensitivity of this method is much better than the centrifugation smear method. The whole process can be accomplished within 30 min. This novel method may have potential as a clinical tool for analysis of bacteria in the CSF.
Collapse
|
8
|
Espinal ER, Sharp SJ, Kim BJ. Induced Pluripotent Stem Cell (iPSC)-Derived Endothelial Cells to Study Bacterial-Brain Endothelial Cell Interactions. Methods Mol Biol 2022; 2492:73-101. [PMID: 35733039 DOI: 10.1007/978-1-0716-2289-6_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bacterial meningitis is a serious infection of the central nervous system (CNS) that occurs when blood-borne bacteria are able to exit the cerebral vasculature and cause inflammation. The blood-brain barrier (BBB) and the meningeal blood-CSF barrier (mBCSFB) are composed of highly specialized brain endothelial cells (BECs) that possess unique phenotypes when compared to their peripheral endothelial counterparts. To cause meningitis, bacterial pathogens must be able to interact and penetrate these specialized BECs to gain access to the CNS. In vitro models have been employed to study bacterial-BEC interactions; however, many lack BEC phenotypes. Induced pluripotent stem cell (iPSC) technologies have enabled the derivation of brain endothelial-like cells that phenocopy BECs in culture. Recently, these iPSC-BECs have been employed to examine the host-pathogen interaction at the endothelial brain barriers. Using two clinically relevant human meningeal pathogens, this chapter describes the use of iPSC-BECs to study various aspects of BEC-bacterial interaction.
Collapse
Affiliation(s)
- Eric R Espinal
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, USA
| | - S Jerod Sharp
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, USA
- Jacksonville State University, Jacksonville, AL, USA
| | - Brandon J Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, USA.
| |
Collapse
|
9
|
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
|
10
|
Herold R, Scholtysik R, Moroniak S, Weiss C, Ishikawa H, Schroten H, Schwerk C. Capsule-dependent impact of MAPK signalling on host cell invasion and immune response during infection of the choroid plexus epithelium by Neisseria meningitidis. Fluids Barriers CNS 2021; 18:53. [PMID: 34863201 PMCID: PMC8643193 DOI: 10.1186/s12987-021-00288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/16/2021] [Indexed: 01/15/2023] Open
Abstract
Background The Gram-negative bacterium Neisseria meningitidis (Nm) can cause meningitis in humans, but the host signalling pathways manipulated by Nm during central nervous system (CNS) entry are not completely understood. Methods We investigate the role of the mitogen-activated protein kinases (MAPK) Erk1/2 and p38 in an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with Nm serogroup B (NmB) and serogroup C (NmC) strains. A transcriptome analysis of HIBCPP cells following infection with Nm by massive analysis of cDNA ends (MACE) was done to further characterize the cellular response to infection of the barrier. Results Interestingly, whereas NmB and NmC wild type strains required active Erk1/2 and p38 pathways for infection, invasion by capsule-deficient mutants was independent of Erk1/2 and, in case of the NmB strain, of p38 activity. The transcriptome analysis of HIBCPP cells following infection with Nm demonstrated specific regulation of genes involved in the immune response dependent on Erk1/2 signalling. Gene ontology (GO) analysis confirmed loss of MAPK signalling after Erk1/2 inhibition and revealed an additional reduction of cellular responses including NFκB and JAK-STAT signalling. Interestingly, GO terms related to TNF signalling and production of IL6 were lost specifically following Erk1/2 inhibition during infection with wild type Nm, which correlated with the reduced infection rates by the wild type in absence of Erk1/2 signalling. Conclusion Our data point towards a role of MAPK signalling during infection of the CP epithelium by Nm, which is strongly influenced by capsule expression, and affects infection rates as well as the host cell response. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00288-7.
Collapse
Affiliation(s)
- Rosanna Herold
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - René Scholtysik
- Genomics & Transcriptomics Facility, Institute of Cell Biology, University Hospital Essen, Virchowstraße 173, 45122, Essen, Germany
| | - Selina Moroniak
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, 1-1-1Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| |
Collapse
|
11
|
Sharma S, Kumar M, Kumar J, Srivastava N, Hussain MA, Shelly A, Mazumder S. M. fortuitum-induced CNS-pathology: Deciphering the role of canonical Wnt signaling, blood brain barrier components and cytokines. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 122:104111. [PMID: 33933535 DOI: 10.1016/j.dci.2021.104111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 06/12/2023]
Abstract
Molecular underpinning of mycobacteria-induced CNS-pathology is not well understood. In the present study, zebrafish were infected with Mycobacterium fortuitum and the prognosis of CNS-pathogenesis studied. We observed M. fortuitum triggers extensive brain-pathology. Evans blue extravasation demonstrated compromised blood-brain barrier (BBB) integrity. Further, decreased expression in tight-junction (TJ) and adherens junction complex (AJC) genes were noted in infected brain. Wnt-signaling has emerged as a major player in host-mycobacterial immunity but its involvement/role in brain-infection is not well studied. Sustained expression of wnt2, wnt3a, fzd5, lrp5/6 and β-catenin, with concordant decline in degradation complex components axin, gsk3β and β-catenin regulator capn2a were observed. The surge in ifng1 and tnfa expression preceding il10 and il4 suggested cytokine-interplay critical in M. fortuitum-induced brain-pathology. Therefore, we suggest adult zebrafish as a viable model for studying CNS-pathology and using the same, conclude that M. fortuitum infection is associated with repressed TJ-AJC gene expression and compromised BBB permeability. Our results implicate Wnt/β-catenin pathway in M. fortuitum-induced CNS-pathology wherein Th1-type signals facilitate bacterial clearance and Th2-type signals prevent the disease sequel.
Collapse
Affiliation(s)
- Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Manmohan Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Nidhi Srivastava
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India; Department of Zoology, School of Basic and Applied Sciences, Maharaja Agrasen University, Solan, Himachal Pradesh, 174103, India
| | - Md Arafat Hussain
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Asha Shelly
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India; Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, 110021, India.
| |
Collapse
|
12
|
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: 101] [Impact Index Per Article: 33.7] [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
|
13
|
Shah KA, Katz JM, Dehdashti AR. Cerebral Abscess After Onyx Embolization of an Arteriovenous Malformation. World Neurosurg 2019; 135:96-99. [PMID: 31841721 DOI: 10.1016/j.wneu.2019.12.009] [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: 10/27/2019] [Revised: 11/30/2019] [Accepted: 12/02/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Endovascular embolization is being increasingly used to treat cerebral arteriovenous malformations. Common complications associated with embolization include intracranial hemorrhage and ischemic stroke; intracranial infections seldomly occur and are even less frequently reported. Although abscess formation after embolization is exceedingly rare, it is a serious condition that warrants immediate attention. CASE DESCRIPTION This 53-year-old male with a ruptured left temporal-occipital arteriovenous malformation. He underwent uncomplicated 2-stage embolization with Onyx and was discharged. Five weeks after embolization, he returned to the hospital with worsening aphasia and contralateral hemiparesis, and a cerebral abscess was detected at the site of embolization. Bacterial cultures were positive for Escherichia coli, and he was treated successfully with surgical excision of the abscess and Onyx material, followed by long-term antibiotics. CONCLUSIONS Although rare, formation of an intracranial abscess after endovascular embolization is a potential complication in the treatment of arteriovenous malformations.
Collapse
Affiliation(s)
- Kevin A Shah
- Department of Neurosurgery, North Shore University Hospital, Manhasset, New York, USA.
| | - Jeffrey M Katz
- Department of Neurology, North Shore University Hospital, Manhasset, New York, USA
| | - Amir R Dehdashti
- Department of Neurosurgery, North Shore University Hospital, Manhasset, New York, USA
| |
Collapse
|
14
|
Meningitic Escherichia coli Induction of ANGPTL4 in Brain Microvascular Endothelial Cells Contributes to Blood-Brain Barrier Disruption via ARHGAP5/RhoA/MYL5 Signaling Cascade. Pathogens 2019; 8:pathogens8040254. [PMID: 31766605 PMCID: PMC6963727 DOI: 10.3390/pathogens8040254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/18/2022] Open
Abstract
Bacterial meningitis is currently recognized as one of the most important life-threatening infections of the central nervous system (CNS) with high morbidity and mortality, despite the advancements in antimicrobial treatment. The disruption of blood–brain barrier (BBB) induced by meningitis bacteria is crucial for the development of bacterial meningitis. However, the complete mechanisms involving in the BBB disruption remain to be elucidated. Here, we found meningitic Escherichia coli induction of angiopoietin-like 4 (ANGPTL4) in brain microvascular endothelial cells (BMECs) contributes to BBB disruption via ARHGAP5/RhoA/MYL5 signaling cascade, by the demonstration that ANGPTL4 was significantly upregulated in meningitis E. coli infection of BMECs as well as mice, and treatment of the recombinant ANGPTL4 protein led to an increased permeability of the BBB in vitro and in vivo. Moreover, we found that ANGPTL4 did not affect the expression of tight junction proteins involved in BBB disruption, but it increased the expression of MYL5, which was found to have a negative role on the regulation of barrier function during meningitic E. coli infection, through the activation of RhoA signaling pathway. To our knowledge, this is the first report demonstrating the disruption of BBB induced by ANGPTL4 through the ARHGAP5/RhoA/MYL5 pathway, which largely supports the involvement of ANGPTL4 during meningitic E. coli invasion and further expands the theoretical basis for the mechanism of bacterial meningitis.
Collapse
|
15
|
Virulence Factors of Meningitis-Causing Bacteria: Enabling Brain Entry across the Blood-Brain Barrier. Int J Mol Sci 2019; 20:ijms20215393. [PMID: 31671896 PMCID: PMC6862235 DOI: 10.3390/ijms20215393] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/16/2022] Open
Abstract
Infections of the central nervous system (CNS) are still a major cause of morbidity and mortality worldwide. Traversal of the barriers protecting the brain by pathogens is a prerequisite for the development of meningitis. Bacteria have developed a variety of different strategies to cross these barriers and reach the CNS. To this end, they use a variety of different virulence factors that enable them to attach to and traverse these barriers. These virulence factors mediate adhesion to and invasion into host cells, intracellular survival, induction of host cell signaling and inflammatory response, and affect barrier function. While some of these mechanisms differ, others are shared by multiple pathogens. Further understanding of these processes, with special emphasis on the difference between the blood-brain barrier and the blood-cerebrospinal fluid barrier, as well as virulence factors used by the pathogens, is still needed.
Collapse
|