1
|
Gowrikumar S, Tarudji A, McDonald BZ, Balusa SS, Kievit FM, Dhawan P. Claudin-1 impairs blood-brain barrier by downregulating endothelial junctional proteins in traumatic brain injury. Tissue Barriers 2025:2470482. [PMID: 40018968 DOI: 10.1080/21688370.2025.2470482] [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: 10/27/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 03/01/2025] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in patients. Brain microvasculature endothelial cells form the blood-brain barrier (BBB) which functions to maintain a protective barrier for the brain from the passive entry of systemic solutes. As a result of the cellular disruption caused by TBI, the BBB is compromised. Tight junction disruption in the endothelium of the BBB has been implicated in this response, but the underlying mechanisms remain unresolved. We utilized various in vivo models of severe to mild TBI as well as in vitro exposure of brain endothelial cells (bEND.3) to analyze conditions encountered following TBI to gain mechanistic insight into alterations observed at the BBB. We found that claudin-1 (CLDN1), was significantly increased in the brain endothelium both in vivo and in vitro. The observed increase of CLDN1 expression correlated with down-regulation of claudin-5 (CLDN5), occludin (OCLN), and zonula occludens (ZO-1), thereby altering BBB integrity by decreasing TEER and increasing permeability. Knockdown of CLDN1 in these pathogenic conditions showed stability of the endothelial junctional proteins. A decline in the epigenetic regulator silent information regulator family protein 1 (SIRT1), a member of the NAD+ dependent protein deacetylases, coincided with this upregulation of CLDN1. Indeed, the quenching of oxidative stress through NAC treatment was able to reduce injury-induced upregulation of CLDN1 in vitro. Mechanistically, an SRC-dependent tyrosine phosphorylation of OCLN and ZO-1 in CLDN1-modulated conditions was observed. Our findings will provide new insights into BBB deregulation and new possible treatment opportunities for TBI.
Collapse
Affiliation(s)
- Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aria Tarudji
- Department of Biological Systems Engineering, University of Nebraska, Lincoln, NE, USA
| | - Brandon Z McDonald
- Department of Biological Systems Engineering, University of Nebraska, Lincoln, NE, USA
| | - Sai Sindhura Balusa
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Forrest M Kievit
- Department of Biological Systems Engineering, University of Nebraska, Lincoln, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
2
|
Wu J, Wang X, Qiao K, Pan N, Chen X, Liu Z, Chen Y. Screening and Antioxidant Activities Evaluation of Peptides From Abalone ( Haliotis discus hannai Ino). Food Sci Nutr 2025; 13:e70028. [PMID: 39926104 PMCID: PMC11807725 DOI: 10.1002/fsn3.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Marine organisms are rich in antioxidant peptides; however, extracting these peptides is time-consuming, labor-intensive, and costly, with sequence losses leading to uncertain results. This study aimed to identify abalone-derived antioxidant peptides with strong Keap1 binding ability and validate their antioxidative activities using a cellular oxidative damage model. We constructed an abalone-derived peptide library comprising 363 peptides using virtual enzymatic hydrolysis techniques. Of the 98 human Keap1 protein structures available in the protein data bank database, 2FLU was selected as the receptor. Using the CDOCKER module in Discovery Studio software, molecular docking was performed with the peptide library as ligands and 2FLU as the receptor, targeting the binding site at coordinates x: 5.000222, y: 7.103889 and z: 5.058000. Ten abalone-derived peptides with the strongest inhibition against Keap1-Nrf2 interaction were identified. A 2,2'-azobis (2-methylpropionamidine) dihydrochloride (AAPH)-induced oxidative damage model in human umbilical vein endothelial cells (HUVECs) was used to verify the molecular docking results and identified DEDEDEDK as the most active antioxidant peptide. DEDEDEDK interferes with Keap1-Nrf2 binding, significantly reducing reactive oxygen species levels in damaged cells, increasing superoxide dismutase and catalase activities, and elevated glutathione content, indicating its potential to mitigate AAPH-induced oxidative damage in HUVECs.
Collapse
Affiliation(s)
- Jingna Wu
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources/Fujian Universities and Colleges Engineering Research Center of Marine Biopharmaceutical ResourcesXiamen Medical CollegeXiamenP. R. China
| | - Xiaoya Wang
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources/Fujian Universities and Colleges Engineering Research Center of Marine Biopharmaceutical ResourcesXiamen Medical CollegeXiamenP. R. China
| | - Kun Qiao
- Fujian Fisheries Research InstituteXiamenP. R. China
| | - Nan Pan
- Fujian Fisheries Research InstituteXiamenP. R. China
| | - Xiaoting Chen
- Fujian Fisheries Research InstituteXiamenP. R. China
| | - Zhiyu Liu
- Fujian Fisheries Research InstituteXiamenP. R. China
| | - Yuanxin Chen
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources/Fujian Universities and Colleges Engineering Research Center of Marine Biopharmaceutical ResourcesXiamen Medical CollegeXiamenP. R. China
| |
Collapse
|
3
|
Mondkar PP, Seo HS, Lodge TP, Azarin SM. Diblock Copolymers of Poly(ethylene oxide)- b-poly(propylene oxide) Stabilize a Blood-Brain Barrier Model under Oxidative Stress. Mol Pharm 2024; 21:5646-5660. [PMID: 39400078 DOI: 10.1021/acs.molpharmaceut.4c00608] [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: 10/15/2024]
Abstract
The blood-brain barrier (BBB) is a highly restrictive barrier at the interface between the brain and the vascular system. Even under BBB dysfunction, it is extremely difficult to deliver therapies across the barrier, limiting the options for treatment of neurological injuries and disorders. To circumvent these challenges, there is interest in developing therapies that directly engage with the damaged BBB to restore its function. Previous studies revealed that poloxamer 188 (P188), a water-soluble triblock copolymer of poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO), partially mitigated BBB dysfunction in vivo. In the context of stabilization of the damaged BBB, the mechanism of action of PEO-PPO block copolymers is unknown, and there has been minimal exploration of polymers beyond P188. In this study, a human-based in vitro BBB model under oxidative stress was used to investigate polymer-BBB interactions since oxidative stress is closely linked with BBB dysfunction in many neurological injuries and disorders. PEO-PPO block copolymers of varied numbers of chemically distinct blocks, PEO block length, and functionality of the end group of the PPO block were assessed for their efficacy in improving key physiological readouts associated with BBB dysfunction. While treatment with P188 did not mitigate damage in the in vitro BBB model, treatment with three diblock copolymers improved barrier integrity under oxidative stress to a similar extent. Of the considered variations in the block copolymer design, the reduction in the number of chemically distinct blocks had the strongest influence on therapeutic function. The demonstrated efficacy of three alternative PEO-PPO diblock copolymers in this work reveals the potential of these polymers as a class of therapeutics that directly treat the damaged BBB, expanding the options for treatment of neurological injuries and disorders.
Collapse
Affiliation(s)
- Pranati P Mondkar
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Hannah S Seo
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Timothy P Lodge
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
McRobb LS, Lee VS, Faqihi F, Stoodley MA. A Simple Model to Study Mosaic Gene Expression in 3D Endothelial Spheroids. J Cardiovasc Dev Dis 2024; 11:305. [PMID: 39452276 PMCID: PMC11508842 DOI: 10.3390/jcdd11100305] [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: 06/07/2024] [Revised: 09/13/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
AIMS The goal of this study was to establish a simple model of 3D endothelial spheroids with mosaic gene expression using adeno-associated virus (AAV) transduction, with a future aim being to study the activity of post-zygotic mutations common to vascular malformations. METHODS In this study, 96-well U-bottom plates coated with a commercial repellent were seeded with two immortalized human endothelial cell lines and aggregation monitored using standard microscopy or live-cell analysis. The eGFP expression was used to monitor the AAV transduction. RESULTS HUVEC-TERT2 could not form spheroids spontaneously. The inclusion of collagen I in the growth medium could stimulate cell aggregation; however, these spheroids were not stable. In contrast, the hCMEC/D3 cells aggregated spontaneously and formed reproducible, robust 3D spheroids within 3 days, growing steadily for at least 4 weeks without the need for media refreshment. The hCMEC/D3 spheroids spontaneously developed a basement membrane, including collagen I, and expressed endothelial-specific CD31 at the spheroid surface. Serotypes AAV1 and AAV2QUADYF transduced these spheroids without toxicity and established sustained, mosaic eGFP expression. CONCLUSIONS In the future, this simple approach to endothelial spheroid formation combined with live-cell imaging could be used to rapidly assess the 3D phenotypes and drug and radiation sensitivities arising from mosaic mutations common to brain vascular malformations.
Collapse
Affiliation(s)
- Lucinda S. McRobb
- Macquarie Medical School, Faculty of Medicine, Health, and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia (M.A.S.)
| | | | | | | |
Collapse
|
5
|
Shabanian K, Shabanian T, Karsai G, Pontiggia L, Paneni F, Ruschitzka F, Beer JH, Saeedi Saravi SS. AQP1 differentially orchestrates endothelial cell senescence. Redox Biol 2024; 76:103317. [PMID: 39180980 PMCID: PMC11388013 DOI: 10.1016/j.redox.2024.103317] [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: 03/18/2024] [Revised: 07/05/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Accumulation of senescent endothelial cells (ECs) with age is a pivotal driver of cardiovascular diseases in aging. However, little is known about the mechanisms and signaling pathways that regulate EC senescence. In this report, we delineate a previously unrecognized role of aquaporin 1 (AQP1) in orchestrating extracellular hydrogen peroxide (H2O2)-induced cellular senescence in aortic ECs. Our findings underscore AQP1's differential impact on senescence hallmarks, including cell-cycle arrest, senescence-associated secretory phenotype (SASP), and DNA damage responses, intricately regulating angiogenesis. In proliferating ECs, AQP1 is crucial for maintaining angiogenic capacity, whereas disruption of AQP1 induces morphological and mitochondrial alterations, culminating in senescence and impaired angiogenesis. Conversely, Aqp1 knockdown or selective blockade of AQP1 in senescent ECs rescues the excess H2O2-induced cellular senescence phenotype and metabolic dysfunction, thereby ameliorating intrinsic angiogenic incompetence. Mechanistically, AQP1 facilitates H2O2 transmembrane transport, exacerbating oxidant-sensitive kinases CaMKII-AMPK. This process suppresses HDAC4 translocation, consequently de-repressing Mef2A-eNOS signaling in proliferating ECs. However, in senescent ECs, AQP1 overexpression is linked to preserved HDAC4-Mef2A complex and downregulation of eNOS signaling. Together, our studies identify AQP1 as a novel epigenetic regulator of HDAC4-Mef2A-dependent EC senescence and angiogenic potential, highlighting its potential as a therapeutic target for antagonizing age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Khatereh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Taraneh Shabanian
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich, 8952, Schlieren, Switzerland
| | - Luca Pontiggia
- Tissue Biology Research Unit, University Children's Hospital Zurich, 8952, Schlieren, Switzerland; Children's Research Center, University Children's Hospital Zurich, 8032, Zurich, Switzerland; Faculty of Medicine, University of Zurich, 8032, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404, Baden, Switzerland.
| | - Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Diao X, Han H, Sun H, Zhang H, Wu W. Protection of Tight Junctional Complexes between hCMEC/D3 Cells by Deep-Sea Fibrinolytic Compound FGFC1. Mar Drugs 2024; 22:341. [PMID: 39195457 DOI: 10.3390/md22080341] [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: 06/15/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Tight junctional complexes (TJCs) between cerebral microvascular endothelial cells (CMECs) are essential parts of the blood-brain barrier (BBB), whose regulation closely correlates to the BBB's integrity and function. hCMEC/D3 is the typical cell line used to imitate and investigate the barrier function of the BBB via the construction of an in vitro model. This study aims to investigate the protective effect of the deep-sea-derived fibrinolytic compound FGFC1 against H2O2-induced dysfunction of TJCs and to elucidate the underlying mechanism. The barrier function was shown to decline following exposure to 1 mM H2O2 in an in vitro model of hCMEC/D3 cells, with a decreasing temperature-corrected transendothelial electrical resistance (tcTEER) value. The decrease in the tcTEER value was significantly inhibited by 80 or 100 µM FGFC1, which suggested it efficiently protected the barrier integrity, allowing it to maintain its function against the H2O2-induced dysfunction. According to immunofluorescence microscopy (IFM) and quantitative real-time polymerase chain reaction (qRT-PCR), compared to the H2O2-treated group, 80~100 µM FGFC1 enhanced the expression of claudin-5 (CLDN-5) and VE-cadherin (VE-cad). And this enhancement was indicated to be mainly achieved by both up-regulation of CLDN-5 and inhibition of the down-regulation by H2O2 of VE-cad at the transcriptional level. Supported by FGFC1's molecular docking to these proteins with reasonable binding energy, FGFC1 was proved to exert a positive effect on TJCs' barrier function in hCMEC/D3 cells via targeting CLDN-5 and VE-cad. This is the first report on the protection against H2O2-induced barrier dysfunction by FGFC1 in addition to its thrombolytic effect. With CLDN-5 and VE-cad as the potential target proteins of FGFC1, this study provides evidence at the cellular and molecular levels for FGFC1's reducing the risk of bleeding transformation following its application in thrombolytic therapy for cerebral thrombosis.
Collapse
Affiliation(s)
- Xiaozhen Diao
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Putuo Sub-Center of International Joint Research Center for Marine Biological Sciences, Zhoushan 316100, China
| | - Hui Han
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Haoyu Sun
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Haixing Zhang
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Wenhui Wu
- Department of Marine Bio-Pharmacology, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai 201306, China
| |
Collapse
|
7
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [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: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
8
|
Ali MA, Gioscia-Ryan R, Yang D, Sutton NR, Tyrrell DJ. Cardiovascular aging: spotlight on mitochondria. Am J Physiol Heart Circ Physiol 2024; 326:H317-H333. [PMID: 38038719 PMCID: PMC11219063 DOI: 10.1152/ajpheart.00632.2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/02/2023]
Abstract
Mitochondria are cellular organelles critical for ATP production and are particularly relevant to cardiovascular diseases including heart failure, atherosclerosis, ischemia-reperfusion injury, and cardiomyopathies. With advancing age, even in the absence of clinical disease, mitochondrial homeostasis becomes disrupted (e.g., redox balance, mitochondrial DNA damage, oxidative metabolism, and mitochondrial quality control). Mitochondrial dysregulation leads to the accumulation of damaged and dysfunctional mitochondria, producing excessive reactive oxygen species and perpetuating mitochondrial dysfunction. In addition, mitochondrial DNA, cardiolipin, and N-formyl peptides are potent activators of cell-intrinsic and -extrinsic inflammatory pathways. These age-related mitochondrial changes contribute to the development of cardiovascular diseases. This review covers the impact of aging on mitochondria and links these mechanisms to therapeutic implications for age-associated cardiovascular diseases.
Collapse
Affiliation(s)
- Md Akkas Ali
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Rachel Gioscia-Ryan
- Department of Anesthesiology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Dongli Yang
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Nadia R Sutton
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, United States
| | - Daniel J Tyrrell
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
9
|
Waithe OY, Peng X, Childs EW, Tharakan B. Measurement of Transendothelial Electrical Resistance in Blood-Brain Barrier Endothelial Cells. Methods Mol Biol 2024; 2711:199-203. [PMID: 37776459 DOI: 10.1007/978-1-0716-3429-5_16] [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: 10/02/2023]
Abstract
The integrity of the blood-brain barrier (BBB), the protective barrier of the brain, is key to maintaining normal microvascular permeability and brain homeostasis. Brain microvascular endothelial cells are primary components of the blood-brain barrier. Transendothelial electrical resistance (TEER) is the electrical resistance across a cellular monolayer such as the brain microvascular endothelial cell monolayers. Measurement of TEER is considered a sensitive, reliable, and noninvasive method for evaluating barrier integrity and permeability of an endothelial cell monolayer under in vitro conditions. Measurement of TEER is also helpful for studying various cellular and molecular changes and signaling events that regulate barrier functions in endothelial monolayers. One of the in vitro endothelial cell barrier models that have been commonly used for measuring TEER is the BBB model using human or rodent brain microvascular endothelial cells grown as a monolayer. In this protocol, we describe how TEER is measured in brain microvascular endothelial cell monolayers, to determine blood-brain barrier integrity under in vitro conditions.
Collapse
Affiliation(s)
- O'lisa Yaa Waithe
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Xu Peng
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, USA
| | - Ed W Childs
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
10
|
Zhao N, Chung TD, Guo Z, Jamieson JJ, Liang L, Linville RM, Pessell AF, Wang L, Searson PC. The influence of physiological and pathological perturbations on blood-brain barrier function. Front Neurosci 2023; 17:1289894. [PMID: 37937070 PMCID: PMC10626523 DOI: 10.3389/fnins.2023.1289894] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
The blood-brain barrier (BBB) is located at the interface between the vascular system and the brain parenchyma, and is responsible for communication with systemic circulation and peripheral tissues. During life, the BBB can be subjected to a wide range of perturbations or stresses that may be endogenous or exogenous, pathological or therapeutic, or intended or unintended. The risk factors for many diseases of the brain are multifactorial and involve perturbations that may occur simultaneously (e.g., two-hit model for Alzheimer's disease) and result in different outcomes. Therefore, it is important to understand the influence of individual perturbations on BBB function in isolation. Here we review the effects of eight perturbations: mechanical forces, temperature, electromagnetic radiation, hypoxia, endogenous factors, exogenous factors, chemical factors, and pathogens. While some perturbations may result in acute or chronic BBB disruption, many are also exploited for diagnostic or therapeutic purposes. The resultant outcome on BBB function depends on the dose (or magnitude) and duration of the perturbation. Homeostasis may be restored by self-repair, for example, via processes such as proliferation of affected cells or angiogenesis to create new vasculature. Transient or sustained BBB dysfunction may result in acute or pathological symptoms, for example, microhemorrhages or hypoperfusion. In more extreme cases, perturbations may lead to cytotoxicity and cell death, for example, through exposure to cytotoxic plaques.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Tracy D. Chung
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - John J. Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Lily Liang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alex F. Pessell
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Linus Wang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
11
|
Maldonado J, Huang JH, Childs EW, Tharakan B. Racial/Ethnic Differences in Traumatic Brain Injury: Pathophysiology, Outcomes, and Future Directions. J Neurotrauma 2023; 40:502-513. [PMID: 36029219 DOI: 10.1089/neu.2021.0455] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability in the United States, exacting a debilitating physical, social, and financial strain. Therefore, it is crucial to examine the impact of TBI on medically underserved communities in the U.S. The purpose of the current study was to review the literature on TBI for evidence of racial/ethnic differences in the U.S. Results of the review showed significant racial/ethnic disparities in TBI outcome and several notable differences in other TBI variables. American Indian/Alaska Natives have the highest rate and number of TBI-related deaths compared with all other racial/ethnic groups; Blacks/African Americans are significantly more likely to incur a TBI from violence when compared with Non-Hispanic Whites; and minorities are significantly more likely to have worse functional outcome compared with Non-Hispanic Whites, particularly among measures of community integration. We were unable to identify any studies that looked directly at underlying racial/ethnic biological variations associated with different TBI outcomes. In the absence of studies on racial/ethnic differences in TBI pathobiology, taking an indirect approach, we looked for studies examining racial/ethnic differences in oxidative stress and inflammation outside the scope of TBI as they are known to heavily influence TBI pathobiology. The literature indicates that Blacks/African Americans have greater inflammation and oxidative stress compared with Non-Hispanic Whites. We propose that future studies investigate the possibility of racial/ethnic differences in inflammation and oxidative stress within the context of TBI to determine whether there is any relationship or impact on TBI outcome.
Collapse
Affiliation(s)
- Justin Maldonado
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott and White Health and Texas A&M University College of Medicine, Temple, Texas, USA
| | - Ed W Childs
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
12
|
Gil E, Wall E, Noursadeghi M, Brown JS. Streptococcus pneumoniae meningitis and the CNS barriers. Front Cell Infect Microbiol 2023; 12:1106596. [PMID: 36683708 PMCID: PMC9845635 DOI: 10.3389/fcimb.2022.1106596] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023] Open
Abstract
Streptococcus pneumoniae (SPN) is a globally significant cause of meningitis, the pathophysiology of which involves damage to the brain by both bacterial virulence factors and the host inflammatory response. In most cases of SPN meningitis bacteria translocate from the blood into the central nervous system (CNS). The principal site of SPN translocation into the CNS is not known, with possible portals of entry proposed to be the cerebral or meningeal blood vessels or the choroid plexus. All require SPN to bind to and translocate across the vascular endothelial barrier, and subsequently the basement membrane and perivascular structures, including an additional epithelial barrier in the case of the blood-CSF barrier. The presence of SPN in the CNS is highly inflammatory resulting in marked neutrophilic infiltration. The secretion of toxic inflammatory mediators by activated neutrophils within the CNS damages pathogen and host alike, including the non-replicative neurons which drives morbidity and mortality. As with the translocation of SPN, the recruitment of neutrophils into the CNS in SPN meningitis necessitates the translocation of neutrophils from the circulation across the vascular barrier, a process that is tightly regulated under basal conditions - a feature of the 'immune specialization' of the CNS. The brain barriers are therefore central to SPN meningitis, both through a failure to exclude bacteria and maintain CNS sterility, and subsequently through the active recruitment and/or failure to exclude circulating leukocytes. The interactions of SPN with these barriers, barrier inflammatory responses, along with their therapeutic implications, are explored in this review.
Collapse
Affiliation(s)
- Eliza Gil
- Division of Infection and Immunity, University College London, London, United Kingdom,*Correspondence: Eliza Gil,
| | - Emma Wall
- Francis Crick Institute, London, United Kingdom,UCLH Biomedical Research Centre, London, United Kingdom
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jeremy S. Brown
- Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
13
|
Sousa T, Gouveia M, Travasso RD, Salvador A. How abundant are superoxide and hydrogen peroxide in the vasculature lumen, how far can they reach? Redox Biol 2022; 58:102527. [PMID: 36335761 PMCID: PMC9640316 DOI: 10.1016/j.redox.2022.102527] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Paracrine superoxide (O2•−) and hydrogen peroxide (H2O2) signaling critically depends on these substances' concentrations, half-lives and transport ranges in extracellular media. Here we estimated these parameters for the lumen of human capillaries, arterioles and arteries using reaction-diffusion-advection models. These models considered O2•− and H2O2 production by endothelial cells and uptake by erythrocytes and endothelial cells, O2•− dismutation, O2•− and H2O2 diffusion and advection by the blood flow. Results show that in this environment O2•− and H2O2 have half-lives <60. ms and <40. ms, respectively, the former determined by the plasma SOD3 activity, the latter by clearance by endothelial cells and erythrocytes. H2O2 concentrations do not exceed the 10 nM scale. Maximal O2•− concentrations near vessel walls exceed H2O2's several-fold when the latter results solely from O2•− dismutation. Cytosolic dismutation of inflowing O2•− may thus significantly contribute to H2O2 delivery to cells. O2•− concentrations near vessel walls decay to 50% of maximum 12 μm downstream from O2•− production sites. H2O2 concentrations in capillaries decay to 50% of maximum 22 μm (6.0 μm) downstream from O2•− (H2O2) production sites. Near arterioles' (arteries') walls, they decay by 50% within 6.0 μm (4. μm) of H2O2 production sites. However, they reach maximal values 50 μm (24 μm) downstream from O2•− production sites and decrease by 50% over 650 μm (500 μm). Arterial/olar endothelial cells might thus signal over a mm downstream through O2•−-derived H2O2, though this requires nM-sensitive H2O2 transduction mechanisms. Physiological local H2O2 concentrations in vasculature lumen are up to 10's of μM. H2O2 transport range in capillaries is just ≈20 μm. Faster blood flow in arteri(ol)es transports O2•−-derived H2O2 over 100's of μm Similar H2O2 abundances and distribution near arterioles' and arteries' walls, likewise for O2•−. Inflowing O2•− may significantly feed H2O2 to the cytosol of endothelial cells
Collapse
|
14
|
Gaire BP. Microglia as the Critical Regulators of Neuroprotection and Functional Recovery in Cerebral Ischemia. Cell Mol Neurobiol 2022; 42:2505-2525. [PMID: 34460037 PMCID: PMC11421653 DOI: 10.1007/s10571-021-01145-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurology and Anesthesiology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
15
|
Tregub PP, Averchuk AS, Baranich TI, Ryazanova MV, Salmina AB. Physiological and Pathological Remodeling of Cerebral Microvessels. Int J Mol Sci 2022; 23:12683. [PMID: 36293539 PMCID: PMC9603917 DOI: 10.3390/ijms232012683] [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: 09/18/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
There is growing evidence that the remodeling of cerebral microvessels plays an important role in plastic changes in the brain associated with development, experience, learning, and memory consolidation. At the same time, abnormal neoangiogenesis, and deregulated regulation of microvascular regression, or pruning, could contribute to the pathogenesis of neurodevelopmental diseases, stroke, and neurodegeneration. Aberrant remodeling of microvesselsis associated with blood-brain barrier breakdown, development of neuroinflammation, inadequate microcirculation in active brain regions, and leads to the dysfunction of the neurovascular unit and progressive neurological deficits. In this review, we summarize current data on the mechanisms of blood vessel regression and pruning in brain plasticity and in Alzheimer's-type neurodegeneration. We discuss some novel approaches to modulating cerebral remodeling and preventing degeneration-coupled aberrant microvascular activity in chronic neurodegeneration.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Federal State Budgetary Scientific Institution Research Center of Neurology, 125367 Moscow, Russia
| | | | | | | | | |
Collapse
|
16
|
Xu Y, Ma Q, Du H, Yang C, Lin G. Postoperative Delirium in Neurosurgical Patients: Recent Insights into the Pathogenesis. Brain Sci 2022; 12:brainsci12101371. [PMID: 36291305 PMCID: PMC9599232 DOI: 10.3390/brainsci12101371] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Postoperative delirium (POD) is a complication characterized by disturbances in attention, awareness, and cognitive function that occur shortly after surgery or emergence from anesthesia. Since it occurs prevalently in neurosurgical patients and poses great threats to the well-being of patients, much emphasis is placed on POD in neurosurgical units. However, there are intricate theories about its pathogenesis and limited pharmacological interventions for POD. In this study, we review the recent insights into its pathogenesis, mainly based on studies within five years, and the five dominant pathological theories that account for the development of POD, with the intention of furthering our understanding and boosting its clinical management.
Collapse
Affiliation(s)
- Yinuo Xu
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Qianquan Ma
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China
- Center for Precision Neurosurgery and Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Haiming Du
- Department of Anesthesiology, Peking University Third Hospital, Beijing 100191, China
| | - Chenlong Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China
- Center for Precision Neurosurgery and Oncology, Peking University Health Science Center, Beijing 100191, China
- North America Medical Education Foundation, Union City, CA 94587, USA
- Correspondence: (C.Y.); (G.L.); Tel.: +86-135-1108-7060 (C.Y.); +86-135-5240-0103 (G.L.)
| | - Guozhong Lin
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China
- Center for Precision Neurosurgery and Oncology, Peking University Health Science Center, Beijing 100191, China
- Correspondence: (C.Y.); (G.L.); Tel.: +86-135-1108-7060 (C.Y.); +86-135-5240-0103 (G.L.)
| |
Collapse
|
17
|
Ju J, Su Y, Zhou Y, Wei H, Xu Q. The SARS-CoV-2 envelope protein disrupts barrier function in an in vitro human blood-brain barrier model. Front Cell Neurosci 2022; 16:897564. [PMID: 36082238 PMCID: PMC9445123 DOI: 10.3389/fncel.2022.897564] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) have been frequently reported to exhibit neurological manifestations and disruption of the blood-brain barrier (BBB). Among the risk factors for BBB breakdown, the loss of endothelial cells and pericytes has caused widespread concern. Recent studies have revealed that severe acute respiratory syndrome coronavirus 2 envelope (S2E) protein caused cell death. We tested the hypothesis that the S2E protein alone could induce BBB dysfunction. The S2E protein bound to human BBB-related cells and inhibited cell viability in a dose- and time-dependent manner. Importantly, the S2E protein disrupted barrier function in an in vitro BBB model composed of HCMEC/D3 (brain endothelial cell line), HBVP (brain vascular pericyte), and U87MG (astrocyte cell line) cells and suppressed the expression of major genes involved in maintaining endothelial permeability and function. In addition, the S2E protein crossed the HCMEC/D3 monolayer. The S2E protein triggered inflammatory responses in HCMEC/D3 and U87MG cells. Taken together, these results show for the first time that the S2E protein has a negative impact on the BBB. Therapies targeting the S2E protein could protect against and treat central nervous system manifestations in COVID-19 patients.
Collapse
Affiliation(s)
- Jiahang Ju
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuwen Su
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - You Zhou
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Wei
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Xu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Qi Xu
| |
Collapse
|
18
|
Santos-Lima B, Pietronigro EC, Terrabuio E, Zenaro E, Constantin G. The role of neutrophils in the dysfunction of central nervous system barriers. Front Aging Neurosci 2022; 14:965169. [PMID: 36034148 PMCID: PMC9404376 DOI: 10.3389/fnagi.2022.965169] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
Leukocyte migration into the central nervous system (CNS) represents a central process in the development of neurological diseases with a detrimental inflammatory component. Infiltrating neutrophils have been detected inside the brain of patients with several neuroinflammatory disorders, including stroke, multiple sclerosis and Alzheimer’s disease. During inflammatory responses, these highly reactive innate immune cells can rapidly extravasate and release a plethora of pro-inflammatory and cytotoxic factors, potentially inducing significant collateral tissue damage. Indeed, several studies have shown that neutrophils promote blood-brain barrier damage and increased vascular permeability during neuroinflammatory diseases. Recent studies have shown that neutrophils migrate into the meninges and choroid plexus, suggesting these cells can also damage the blood-cerebrospinal fluid barrier (BCSFB). In this review, we discuss the emerging role of neutrophils in the dysfunction of brain barriers across different neuroinflammatory conditions and describe the molecular basis and cellular interplays involved in neutrophil-mediated injury of the CNS borders.
Collapse
|
19
|
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
|
20
|
Rudd H, Toborek M. Pitfalls of Antiretroviral Therapy: Current Status and Long-Term CNS Toxicity. Biomolecules 2022; 12:biom12070894. [PMID: 35883450 PMCID: PMC9312798 DOI: 10.3390/biom12070894] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 02/04/2023] Open
Abstract
HIV can traverse the BBB using a Trojan horse-like mechanism. Hidden within infected immune cells, HIV can infiltrate the highly safeguarded CNS and propagate disease. Once integrated within the host genome, HIV becomes a stable provirus, which can remain dormant, evade detection by the immune system or antiretroviral therapy (ART), and result in rebound viraemia. As ART targets actively replicating HIV, has low BBB penetrance, and exposes patients to long-term toxicity, further investigation into novel therapeutic approaches is required. Viral proteins can be produced by latent HIV, which may play a synergistic role alongside ART in promoting neuroinflammatory pathophysiology. It is believed that the ability to specifically target these proviral reservoirs would be a vital driving force towards a cure for HIV infection. A novel drug design platform, using the in-tandem administration of several therapeutic approaches, can be used to precisely target the various components of HIV infection, ultimately leading to the eradication of active and latent HIV and a functional cure for HIV. The aim of this review is to explore the pitfalls of ART and potential novel therapeutic alternatives.
Collapse
Affiliation(s)
- Harrison Rudd
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065 Katowice, Poland
- Correspondence: ; Tel.: +1-(305)-243-0230
| |
Collapse
|
21
|
Chung TD, Linville RM, Guo Z, Ye R, Jha R, Grifno GN, Searson PC. Effects of acute and chronic oxidative stress on the blood-brain barrier in 2D and 3D in vitro models. Fluids Barriers CNS 2022; 19:33. [PMID: 35551622 PMCID: PMC9097350 DOI: 10.1186/s12987-022-00327-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is a shared pathology of neurodegenerative disease and brain injuries, and is derived from perturbations to normal cell processes by aging or environmental factors such as UV exposure and air pollution. As oxidative cues are often present in systemic circulation, the blood-brain barrier (BBB) plays a key role in mediating the effect of these cues on brain dysfunction. Therefore, oxidative damage and disruption of the BBB is an emergent focus of neurodegenerative disease etiology and progression. We assessed barrier dysfunction in response to chronic and acute oxidative stress in 2D and 3D in vitro models of the BBB with human iPSC-derived brain microvascular endothelial-like cells (iBMECs). We first established doses of hydrogen peroxide to induce chronic damage (modeling aging and neurodegenerative disease) and acute damage (modeling the response to traumatic brain injury) by assessing barrier function via transendothelial electrical resistance in 2D iBMEC monolayers and permeability and monolayer integrity in 3D tissue-engineered iBMEC microvessels. Following application of these chronic and acute doses in our in vitro models, we found local, discrete structural changes were the most prevalent responses (rather than global barrier loss). Additionally, we validated unique functional changes in response to oxidative stress, including dysfunctional cell turnover dynamics and immune cell adhesion that were consistent with changes in gene expression.
Collapse
Affiliation(s)
- Tracy D Chung
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
| | - Robert Ye
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Ria Jha
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gabrielle N Grifno
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, 100 Croft Hall, 3400 North Charles Street, 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
|
22
|
Ayesha A, Bahnson EM, Kayashima Y, Wilder J, Huynh PK, Hiller S, Maeda-Smithies N, Li F. Vitamin B12 does not increase cell viability after hydrogen peroxide induced damage in mouse kidney proximal tubular cells and brain endothelial cells. ADVANCES IN REDOX RESEARCH 2022; 4:100029. [PMID: 35515703 PMCID: PMC9067605 DOI: 10.1016/j.arres.2022.100029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Vitamin B12 (B12) is an essential co-factor for two enzymes in mammalian metabolism and can also act as a mimetic of superoxide dismutase (SOD) converting superoxide (O2 •‒) to hydrogen peroxide (H2O2). High oral dose B12 decreases renal O2 •‒ and post-ischemia/reperfusion injury in mice and protects against damage induced by hypoxia/reperfusion in mouse kidney proximal tubular cells (BU.MPT). O2 •‒ is unstable and rapidly converted to H2O2. H2O2 mediates oxidative stress associated with O2 •‒. Whether B12 protects against damage induced by H2O2 is unknown. Both BU.MPT cells and mouse brain endothelial cells (bEdn.3) were applied to test the effects of B12 on H2O2-induced cytotoxicity. Both types of cells were treated with different doses of H2O2 with or without different doses of B12. Cell viability was analyzed 24 h later. H2O2 caused cell death only at a very high dose, and high pharmacological dose of B12 did not prevent this detrimental effect in either cell type. In bEnd.3 cells, transcriptional levels of heme oxygenase-1 (HO-1) increased, while nuclear factor erythroid 2-related factor 2 (Nrf2) decreased by H2O2. The levels of transcripts were not affected by the B12 treatment. We conclude that the cytotoxic effects of exogenous H2O2 in BU.MPT and bEdn.3 cells are not prevented by B12.
Collapse
Affiliation(s)
- Azraa Ayesha
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 703 Brinkhous-Bullitt Bldg, CB # 7525, Chapel Hill, NC 27599, USA
| | - Edward M Bahnson
- Department of Cell Biology and Physiology, The University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yukako Kayashima
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 703 Brinkhous-Bullitt Bldg, CB # 7525, Chapel Hill, NC 27599, USA
| | - Jennifer Wilder
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 703 Brinkhous-Bullitt Bldg, CB # 7525, Chapel Hill, NC 27599, USA
| | - Phillip K Huynh
- Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Sylvia Hiller
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 703 Brinkhous-Bullitt Bldg, CB # 7525, Chapel Hill, NC 27599, USA
| | - Nobuyo Maeda-Smithies
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 703 Brinkhous-Bullitt Bldg, CB # 7525, Chapel Hill, NC 27599, USA
| | - Feng Li
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 703 Brinkhous-Bullitt Bldg, CB # 7525, Chapel Hill, NC 27599, USA
| |
Collapse
|
23
|
van Leeuwen E, Hampton MB, Smyth LCD. Hypothiocyanous Acid Disrupts the Barrier Function of Brain Endothelial Cells. Antioxidants (Basel) 2022; 11:antiox11040608. [PMID: 35453292 PMCID: PMC9030776 DOI: 10.3390/antiox11040608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a common feature of neurological diseases. During neuroinflammation, neutrophils are recruited to the brain vasculature, where myeloperoxidase can produce hypochlorous acid and the less well-studied oxidant hypothiocyanous acid (HOSCN). In this study, we exposed primary brain endothelial cells (BECs) to HOSCN and observed a rapid loss of transendothelial electrical resistance (TEER) at sublethal concentrations. Decreased barrier function was associated with a loss of tight junctions at cellular contacts and a concomitant loss of dynamic microtubules. Both tight junction and cytoskeletal disruptions were visible within 30 min of exposure, whereas significant loss of TEER took more than 1 h. The removal of the HOSCN after 30 min prevented subsequent barrier dysfunction. These results indicate that BECs are sensitive to HOSCN, resulting in the eventual loss of barrier function. We hypothesise that this mechanism may be relevant in neutrophil transmigration, with HOSCN facilitating blood–brain barrier opening at the sites of egress. Furthermore, this mechanism may be a way through which neutrophils, residing in the vasculature, can influence neuroinflammation in diseases.
Collapse
Affiliation(s)
- Eveline van Leeuwen
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (E.v.L.); (M.B.H.)
| | - Mark B. Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (E.v.L.); (M.B.H.)
| | - Leon C. D. Smyth
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8011, New Zealand; (E.v.L.); (M.B.H.)
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
- Correspondence: ; Tel.: +64-3-378-6225
| |
Collapse
|
24
|
Malard E, Valable S, Bernaudin M, Pérès E, Chatre L. The Reactive Species Interactome in the Brain. Antioxid Redox Signal 2021; 35:1176-1206. [PMID: 34498917 DOI: 10.1089/ars.2020.8238] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significance: Redox pioneer Helmut Sies attempted to explain reactive species' challenges faced by organelles, cells, tissues, and organs via three complementary definitions: (i) oxidative stress, that is, the disturbance in the prooxidant-antioxidant defense balance in favor of the prooxidants; (ii) oxidative eustress, the low physiological exposure to prooxidants; and (iii) oxidative distress, the supraphysiological exposure to prooxidants. Recent Advances: Identification, concentration, and interactions are the most important elements to improve our understanding of reactive species in physiology and pathology. In this context, the reactive species interactome (RSI) is a new multilevel redox regulatory system that identifies reactive species families, reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species, and it integrates their interactions with their downstream biological targets. Critical Issues: We propose a united view to fully combine reactive species identification, oxidative eustress and distress, and the RSI system. In this view, we also propose including the forgotten reactive carbonyl species, an increasingly rediscovered reactive species family related to the other reactive families, and key enzymes within the RSI. We focus on brain physiology and pathology to demonstrate why this united view should be considered. Future Directions: More studies are needed for an improved understanding of the contributions of reactive species through their identification, concentration, and interactions, including in the brain. Appreciating the RSI in its entirety should unveil new molecular players and mechanisms in physiology and pathology in the brain and elsewhere.
Collapse
Affiliation(s)
- Elise Malard
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Myriam Bernaudin
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Elodie Pérès
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| | - Laurent Chatre
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy group, GIP Cyceron, Caen, France
| |
Collapse
|
25
|
The Blood-Brain Barrier, Oxidative Stress, and Insulin Resistance. Antioxidants (Basel) 2021; 10:antiox10111695. [PMID: 34829566 PMCID: PMC8615183 DOI: 10.3390/antiox10111695] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) is a network of specialized endothelial cells that regulates substrate entry into the central nervous system (CNS). Acting as the interface between the periphery and the CNS, the BBB must be equipped to defend against oxidative stress and other free radicals generated in the periphery to protect the CNS. There are unique features of brain endothelial cells that increase the susceptibility of these cells to oxidative stress. Insulin signaling can be impacted by varying levels of oxidative stress, with low levels of oxidative stress being necessary for signaling and higher levels being detrimental. Insulin must cross the BBB in order to access the CNS, levels of which are important in peripheral metabolism as well as cognition. Any alterations in BBB transport due to oxidative stress at the BBB could have downstream disease implications. In this review, we cover the interactions of oxidative stress at the BBB, how insulin signaling is related to oxidative stress, and the impact of the BBB in two diseases greatly affected by oxidative stress and insulin resistance: diabetes mellitus and Alzheimer’s disease.
Collapse
|
26
|
Negri S, Faris P, Moccia F. Reactive Oxygen Species and Endothelial Ca 2+ Signaling: Brothers in Arms or Partners in Crime? Int J Mol Sci 2021; 22:ijms22189821. [PMID: 34575985 PMCID: PMC8465413 DOI: 10.3390/ijms22189821] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
An increase in intracellular Ca2+ concentration ([Ca2+]i) controls virtually all endothelial cell functions and is, therefore, crucial to maintain cardiovascular homeostasis. An aberrant elevation in endothelial can indeed lead to severe cardiovascular disorders. Likewise, moderate amounts of reactive oxygen species (ROS) induce intracellular Ca2+ signals to regulate vascular functions, while excessive ROS production may exploit dysregulated Ca2+ dynamics to induce endothelial injury. Herein, we survey how ROS induce endothelial Ca2+ signals to regulate vascular functions and, vice versa, how aberrant ROS generation may exploit the Ca2+ handling machinery to promote endothelial dysfunction. ROS elicit endothelial Ca2+ signals by regulating inositol-1,4,5-trisphosphate receptors, sarco-endoplasmic reticulum Ca2+-ATPase 2B, two-pore channels, store-operated Ca2+ entry (SOCE), and multiple isoforms of transient receptor potential (TRP) channels. ROS-induced endothelial Ca2+ signals regulate endothelial permeability, angiogenesis, and generation of vasorelaxing mediators and can be exploited to induce therapeutic angiogenesis, rescue neurovascular coupling, and induce cancer regression. However, an increase in endothelial [Ca2+]i induced by aberrant ROS formation may result in endothelial dysfunction, inflammatory diseases, metabolic disorders, and pulmonary artery hypertension. This information could pave the way to design alternative treatments to interfere with the life-threatening interconnection between endothelial ROS and Ca2+ signaling under multiple pathological conditions.
Collapse
|
27
|
Akhter MS, Barabutis N. Suppression of reactive oxygen species in endothelial cells by an antagonist of growth hormone-releasing hormone. J Biochem Mol Toxicol 2021; 35:e22879. [PMID: 34369038 DOI: 10.1002/jbt.22879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/16/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Growth hormone-releasing hormone (GHRH) is a hypothalamic hormone, which regulates the secretion of growth hormone (GH) from the anterior pituitary gland. The effects of GHRH extend beyond the GH-insulin-like growth factor I axis, and that neuropeptide has been involved in the potentiation of several malignancies and other inflammatory disorders. The development of GHRH antagonists (GHRHAnt) delivers an exciting possibility to counteract the pathogenesis of the GHRH-related effects in human pathophysiology, especially when considered that GHRHAnt support endothelial barrier integrity. Those GHRHAnt-mediated effects are exerted at least in part due to the suppression of major inflammatory pathways, and the modulation of major cytoskeletal components. In the present study, we measured the production of reactive oxygen species (ROS) in bovine pulmonary artery endothelial cells, human cerebral microvascular endothelial cells, and human lung microvascular endothelial cells exposed to GHRH or a commercially available GHRHAnt. Our findings reveal the antioxidative effects of GHRHAnt in all three cell lines, which express GHRH receptors. The redox status of NIH/3T3 cells, which do not produce GHRH receptors, was not significantly affected by GHRH or GHRHAnt. Hence, the application of GHRHAnt in pathologies related to increased ROS production should be further investigated.
Collapse
Affiliation(s)
- Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| |
Collapse
|
28
|
Augustine R, Aqel AH, Kalva SN, Joshy KS, Nayeem A, Hasan A. Bioengineered microfluidic blood-brain barrier models in oncology research. Transl Oncol 2021; 14:101087. [PMID: 33865030 PMCID: PMC8066424 DOI: 10.1016/j.tranon.2021.101087] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/25/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Metastasis is the major reason for most brain tumors with up to a 50% chance of occurrence in patients with other types of malignancies. Brain metastasis occurs if cancer cells succeed to cross the 'blood-brain barrier' (BBB). Moreover, changes in the structure and function of BBB can lead to the onset and progression of diseases including neurological disorders and brain-metastases. Generating BBB models with structural and functional features of intact BBB is highly important to better understand the molecular mechanism of such ailments and finding novel therapeutic agents targeting them. Hence, researchers are developing novel in vitro BBB platforms that can recapitulate the structural and functional characteristics of BBB. Brain endothelial cells-based in vitro BBB models have thus been developed to investigate the mechanism of brain metastasis through BBB and facilitate the testing of brain targeted anticancer drugs. Bioengineered constructs integrated with microfluidic platforms are vital tools for recapitulating the features of BBB in vitro closely as possible. In this review, we outline the fundamentals of BBB biology, recent developments in the microfluidic BBB platforms, and provide a concise discussion of diverse types of bioengineered BBB models with an emphasis on the application of them in brain metastasis and cancer research in general. We also provide insights into the challenges and prospects of the current bioengineered microfluidic platforms in cancer research.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| | - Ahmad H Aqel
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Sumama Nuthana Kalva
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - K S Joshy
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar
| | - Ajisha Nayeem
- Department of Biotechnology, St. Mary's College, Thrissur 680020, Kerala, India
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713 Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713 Doha, Qatar.
| |
Collapse
|
29
|
Liu CD, Liu NN, Zhang S, Ma GD, Yang HG, Kong LL, Du GH. Salvianolic acid A prevented cerebrovascular endothelial injury caused by acute ischemic stroke through inhibiting the Src signaling pathway. Acta Pharmacol Sin 2021; 42:370-381. [PMID: 33303991 PMCID: PMC8027612 DOI: 10.1038/s41401-020-00568-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/29/2020] [Indexed: 12/27/2022]
Abstract
Stroke is an acute cerebrovascular disease caused by ruptured or blocked blood vessels. For the prevention of ischemic stroke, the coagulation state of blood and cerebrovascular protection should be considered. Our previous study has shown that salvianolic acid A (SAA), which is a water-soluble component from the root of Salvia Miltiorrhiza Bge, prevents thrombosis with a mild inhibitory effect on platelet aggregation. In this study we investigated the preventive effects of SAA on cerebrovascular endothelial injury caused by ischemia in vivo and oxygen-glucose deprivation (OGD) in vitro, and explored the underlying mechanisms. An autologous thrombus stroke model was established in SD rats by electrocoagulation. SAA (10 mg/kg) was orally administered twice a day for 5 days before the operation. The rats were sacrificed at 24 h after the operation. We showed that pretreatment with SAA significantly improved the neurological deficits, intracerebral hemorrhage, BBB disruption, and vascular endothelial dysfunction as compared with model group. In human brain microvascular endothelial cells (HBMECs), pretreatment with SAA (10 μM) significantly inhibited OGD-induced cell viability reduction and degradation of tight junction proteins (ZO-1, occludin, claudin-5). Furthermore, we found that SAA inhibited the upregulation of Src signaling pathway in vivo and vitro and reversed the increased expression of matrix metalloproteinases (MMPs) after ischemic stroke. In conclusion, our results suggest that SAA protects cerebrovascular endothelial cells against ischemia and OGD injury via suppressing Src signaling pathway. These findings show that pretreatment with SAA is a potential therapeutic strategy for the prevention of ischemic stroke.
Collapse
Affiliation(s)
- Cheng-di Liu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nan-Nan Liu
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Sen Zhang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Guo-Dong Ma
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hai-Guang Yang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ling-Lei Kong
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guan-Hua Du
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
30
|
Santiago Santana JM, Vega-Torres JD, Ontiveros-Angel P, Bin Lee J, Arroyo Torres Y, Cruz Gonzalez AY, Aponte Boria E, Zabala Ortiz D, Alvarez Carmona C, Figueroa JD. Oxidative stress and neuroinflammation in a rat model of co-morbid obesity and psychogenic stress. Behav Brain Res 2021; 400:112995. [PMID: 33301815 PMCID: PMC8713435 DOI: 10.1016/j.bbr.2020.112995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/28/2020] [Accepted: 10/29/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND There is growing recognition for a reciprocal, bidirectional link between anxiety disorders and obesity. Although the mechanisms linking obesity and anxiety remain speculative, this bidirectionality suggests shared pathophysiological processes. Neuroinflammation and oxidative damage are implicated in both pathological anxiety and obesity. This study investigates the relative contribution of comorbid diet-induced obesity and stress-induced anxiety to neuroinflammation and oxidative stress. METHODS Thirty-six (36) male Lewis rats were divided into four groups based on diet type and stress exposure: 1) control diet unexposed (CDU) and 2) exposed (CDE), 3) Western-like high-saturated fat diet unexposed (WDU) and 4) exposed (WDE). Neurobehavioral tests were performed to assess anxiety-like behaviors. The catalytic concentrations of glutathione peroxidase and reductase were measured from plasma samples, and neuroinflammatory/oxidative stress biomarkers were measured from brain samples using Western blot. Correlations between behavioral phenotypes and biomarkers were assessed with Pearson's correlation procedures. RESULTS We found that WDE rats exhibited markedly increased levels of glial fibrillary acidic protein (185 %), catalase protein (215 %), and glutathione reductase (GSHR) enzymatic activity (418 %) relative to CDU rats. Interestingly, the brain protein levels of glutathione peroxidase (GPx) and catalase were positively associated with body weight and behavioral indices of anxiety. CONCLUSIONS Together, our results support a role for neuroinflammation and oxidative stress in heightened emotional reactivity to obesogenic environments and psychogenic stress. Uncovering adaptive responses to obesogenic environments characterized by high access to high-saturated fat/high-sugar diets and toxic stress has the potential to strongly impact how we treat psychiatric disorders in at-risk populations.
Collapse
Affiliation(s)
- Jose M Santiago Santana
- Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, University of Puerto Rico Carolina Campus, Puerto Rico
| | - Julio D Vega-Torres
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Perla Ontiveros-Angel
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jeong Bin Lee
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Yaria Arroyo Torres
- Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, University of Puerto Rico Carolina Campus, Puerto Rico; Universidad Metropolitana de Cupey Sciences and Technology School, Puerto Rico
| | - Alondra Y Cruz Gonzalez
- Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, University of Puerto Rico Carolina Campus, Puerto Rico
| | - Esther Aponte Boria
- Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, University of Puerto Rico Carolina Campus, Puerto Rico
| | - Deisha Zabala Ortiz
- Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, University of Puerto Rico Carolina Campus, Puerto Rico
| | - Carolina Alvarez Carmona
- Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, University of Puerto Rico Carolina Campus, Puerto Rico
| | - Johnny D Figueroa
- Center for Health Disparities and Molecular Medicine and Department of Basic Sciences, Physiology Division, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States.
| |
Collapse
|
31
|
Uddin MA, Akhter MS, Kubra KT, Whitaker KE, Shipley SL, Smith LM, Barabutis N. Hsp90 inhibition protects the brain microvascular endothelium against oxidative stress. BRAIN DISORDERS 2021; 1. [PMID: 33569547 PMCID: PMC7869856 DOI: 10.1016/j.dscb.2020.100001] [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] [Indexed: 12/15/2022] Open
Abstract
The brain endothelium is an integral element of the blood-brain barrier (BBB). Dysfunction of this formation due to increased generation of reactive oxygen species (ROS) progresses the establishment of neurological disorders including stroke and traumatic brain injury. Heat shock protein 90 inhibitors are anti-inflammatory agents, and their activities are mediated, at least in part, by P53. This is a tumor suppressor protein which regulates the opposing activities of Rac1 and RhoA in the cellular cytoskeleton. In the present study we investigated the role of Hsp90 inhibitors in the H2O2-induced brain endothelium breakdown, by employing human cerebral microvascular endothelial cells (hCMEC/D3). Our findings suggest that H2O2 downregulates P53 by enhancing the P53 suppressor mouse double minute 2 homolog (MDM2), as well as by increasing the apyrimidinic endonuclease 1/redox factor 1 (APE1/Ref1). The H2O2 – triggered violation of the brain endothelium barrier was reflected in measurements of transendothelial resistance, and the increased expression of the key cytoskeletal modulators cofilin and myosin light chain 2 (MLC2). Treatment of the hCMEC/D3 cells with Hsp90 inhibitors counteracted those events, and reduced the generation of the hydrogen peroxide – induced reactive oxygen species. Hence, our study suggests that Hsp90 inhibition supports the BBB integrity, and may represent a promising therapeutic approach for disorders associated with brain endothelium breakdown; including COVID-19.
Collapse
Affiliation(s)
- Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Kathryn E Whitaker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Summer L Shipley
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Landon M Smith
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA 71201, USA
| |
Collapse
|
32
|
Abe RJ, Savage H, Imanishi M, Banerjee P, Kotla S, Paez-Mayorga J, Taunton J, Fujiwara K, Won JH, Yusuf SW, Palaskas NL, Banchs J, Lin SH, Schadler KL, Abe JI, Le NT. p90RSK-MAGI1 Module Controls Endothelial Permeability by Post-translational Modifications of MAGI1 and Hippo Pathway. Front Cardiovasc Med 2020; 7:542485. [PMID: 33304925 PMCID: PMC7693647 DOI: 10.3389/fcvm.2020.542485] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 10/15/2020] [Indexed: 01/05/2023] Open
Abstract
Previously, we reported that post-translational modifications (PTMs) of MAGI1, including S741 phosphorylation and K931 de-SUMOylation, both of which are regulated by p90RSK activation, lead to endothelial cell (EC) activation. However, roles for p90RSK and MAGI1-PTMs in regulating EC permeability remain unclear despite MAGI1 being a junctional molecule. Here, we show that thrombin (Thb)-induced EC permeability, detected by the electric cell-substrate impedance sensing (ECIS) based system, was decreased by overexpression of dominant negative p90RSK or a MAGI1-S741A phosphorylation mutant, but was accelerated by overexpression of p90RSK, siRNA-mediated knockdown of magi1, or the MAGI1-K931R SUMOylation mutant. MAGI1 depletion also increased the mRNA and protein expression of the large tumor suppressor kinases 1 and 2 (LATS1/2), which inhibited YAP/TAZ activity and increased EC permeability. Because the endothelial barrier is a critical mediator of tumor hypoxia, we also evaluated the role of p90RSK activation in tumor vessel leakiness by using a relatively low dose of the p90RSK specific inhibitor, FMK-MEA. FMK-MEA significantly inhibited tumor vessel leakiness at a dose that does not affect morphology and growth of tumor vessels in vivo. These results provide novel insights into crucial roles for p90RSK-mediated MAGI1 PTMs and the Hippo pathway in EC permeability, as well as p90RSK activation in tumor vessel leakiness.
Collapse
Affiliation(s)
- Rei J Abe
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Hannah Savage
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Priyanka Banerjee
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jesus Paez-Mayorga
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jong Hak Won
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas L Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jose Banchs
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Keri L Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
33
|
Linville RM, Arevalo D, Maressa JC, Zhao N, Searson PC. Three-dimensional induced pluripotent stem-cell models of human brain angiogenesis. Microvasc Res 2020; 132:104042. [PMID: 32673611 DOI: 10.1016/j.mvr.2020.104042] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022]
Abstract
During brain development, chemical cues released by developing neurons, cellular signaling with pericytes, and mechanical cues within the brain extracellular matrix (ECM) promote angiogenesis of brain microvascular endothelial cells (BMECs). Angiogenesis is also associated with diseases of the brain due to pathological chemical, cellular, and mechanical signaling. Existing in vitro and in vivo models of brain angiogenesis have key limitations. Here, we develop a high-throughput in vitro blood-brain barrier (BBB) bead assay of brain angiogenesis utilizing 150 μm diameter beads coated with induced pluripotent stem-cell (iPSC)-derived human BMECs (dhBMECs). After embedding the beads within a 3D matrix, we introduce various chemical cues and extracellular matrix components to explore their effects on angiogenic behavior. Based on the results from the bead assay, we generate a multi-scale model of the human cerebrovasculature within perfusable three-dimensional tissue-engineered blood-brain barrier microvessels. A sprouting phenotype is optimized in confluent monolayers of dhBMECs using chemical treatment with vascular endothelial growth factor (VEGF) and wnt ligands, and the inclusion of pro-angiogenic ECM components. As a proof-of-principle that the bead angiogenesis assay can be applied to study pathological angiogenesis, we show that oxidative stress can exert concentration-dependent effects on angiogenesis. Finally, we demonstrate the formation of a hierarchical microvascular model of the human blood-brain barrier displaying key structural hallmarks. We develop two in vitro models of brain angiogenesis: the BBB bead assay and the tissue-engineered BBB microvessel model. These platforms provide a tool kit for studies of physiological and pathological brain angiogenesis, with key advantages over existing two-dimensional models.
Collapse
Affiliation(s)
- Raleigh M Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States of America; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States of America
| | - Diego Arevalo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States of America; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States of America
| | - Joanna C Maressa
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States of America; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States of America
| | - Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States of America
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States of America; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States of America; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States of America.
| |
Collapse
|
34
|
Mészáros Á, Molnár K, Nógrádi B, Hernádi Z, Nyúl-Tóth Á, Wilhelm I, Krizbai IA. Neurovascular Inflammaging in Health and Disease. Cells 2020; 9:cells9071614. [PMID: 32635451 PMCID: PMC7407516 DOI: 10.3390/cells9071614] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/02/2020] [Indexed: 12/19/2022] Open
Abstract
Aging is characterized by a chronic low-grade sterile inflammation dubbed as inflammaging, which in part originates from accumulating cellular debris. These, acting as danger signals with many intrinsic factors such as cytokines, are sensed by a network of pattern recognition receptors and other cognate receptors, leading to the activation of inflammasomes. Due to the inflammasome activity-dependent increase in the levels of pro-inflammatory interleukins (IL-1β, IL-18), inflammation is initiated, resulting in tissue injury in various organs, the brain and the spinal cord included. Similarly, in age-related diseases of the central nervous system (CNS), inflammasome activation is a prominent moment, in which cells of the neurovascular unit occupy a significant position. In this review, we discuss the inflammatory changes in normal aging and summarize the current knowledge on the role of inflammasomes and contributing mechanisms in common CNS diseases, namely Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis and stroke, all of which occur more frequently with aging.
Collapse
Affiliation(s)
- Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Doctoral School of Biology, University of Szeged, 6726 Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Theoretical Medicine Doctoral School, University of Szeged, 6720 Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Zsófia Hernádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
| | - István A. Krizbai
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
- Correspondence: ; Tel.: +36-62-599-794
| |
Collapse
|
35
|
Redox signalling and regulation of the blood-brain barrier. Int J Biochem Cell Biol 2020; 125:105794. [PMID: 32562769 DOI: 10.1016/j.biocel.2020.105794] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
Neurological disorders are associated with increased oxidative stress. Reactive oxidants damage tissue and promote cell death, but it is apparent that oxidants can have more subtle effects on cell function through the modulation of redox-sensitive signalling pathways. Cells of the blood-brain barrier regulate the brain microenvironment but become dysfunctional during neurological disease. The blood-brain barrier is maintained by many cell types, and is modulated by redox-sensitive pathways, ranging from the cytoskeletal elements responsible for establishing a barrier, to growth factor and cytokine signalling pathways that influence neurovascular cells. During neurological disease, blood-brain barrier cells are exposed to exogenously generated oxidants from immune cells, as well as increasing endogenously oxidant production. These oxidants impair the function of the blood-brain barrier, leading to increased leakage and reduced blood flow. Reducing the impact of oxidants on the function of blood-brain barrier cells may provide new strategies for delaying the progression of neurological disease.
Collapse
|
36
|
Oxidative stress facilitates cell death by inhibiting Orai1-mediated Ca 2+ entry in brain capillary endothelial cells. Biochem Biophys Res Commun 2019; 523:153-158. [PMID: 31839216 DOI: 10.1016/j.bbrc.2019.12.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022]
Abstract
Brain capillary endothelial cells (BCECs) form the blood-brain barrier (BBB) and play an essential role in the regulation of its functions. Oxidative stress accumulates excessive reactive oxygen species (ROS) and facilitates the death of BCECs, leading to a dysfunctional BBB. However, the mechanisms underlying the death of BCECs under oxidative stress remain unclear. In the present study, the effects of oxidative stress on cell viability, ROS production, intracellular Ca2+ concentration, and protein expression were examined using a cell line derived from bovine BCECs, t-BBEC117. When t-BBEC117 cells were exposed to oxidative stress induced by hydrogen peroxide (H2O2, 10-100 μM), cell growth was inhibited in a dose-dependent manner. Oxidative stress by 30 μM H2O2 increased the production of ROS and its effects were blocked by the ROS scavenger, 10 mM N-acetyl-l-cysteine (NAC). In addition, oxidative stress reduced store-operated Ca2+ entry (SOCE) and this decrease was recovered by NAC or the Orai channel activator, 5 μM 2-aminoethyl diphenylborinate (2-APB). The siRNA knockdown of Orai1 revealed that Orai1 was mainly responsible for SOCE channels and its activity was decreased by oxidative stress. However, the protein expression of Orai1 and STIM1 was not affected by oxidative stress. Oxidative stress-induced cell death was rescued by 2-APB, NAC, or the STIM-Orai activating region. In conclusion, oxidative stress reduces Orai1-mediated SOCE and, thus, facilitates the death of BCECs.
Collapse
|
37
|
Antioxidant properties of probucol released from mesoporous silica. Eur J Pharm Sci 2019; 138:105038. [PMID: 31398394 DOI: 10.1016/j.ejps.2019.105038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/15/2019] [Accepted: 08/05/2019] [Indexed: 11/21/2022]
Abstract
Antioxidants play a vital role in scavenging reactive oxygen species (ROS) produced by the reduction of molecular oxygen from various cellular mechanisms. Under oxidative stress, an increase in the levels of ROS overwhelms the antioxidant response, causing oxidative damage to biological molecules, and leading to the development of various diseases. Drug compounds with potent antioxidant properties are typically poorly water soluble and highly hydrophobic. An extreme case is Probucol (PB), a potent antioxidant with reported water solubility of 5 ng/ml, and oral bioavailiability of <10%. In this study, PB was loaded in mesoporous silica at various drug loadings to understand the changes to the physical properties of the loaded drug, and it's in vitro drug release. Further in vitro studies were conducted in endothelial and microglia cell models to compare the free radical scavening efficiency of ascorbic acid, PB, and PB release from mesoporous silica particles. Out of the three different mesostructured particles studied, the maximum loading of PB was achieved for large pore mesoporous particles (SBA-15) at 50 wt% drug loading, before complete pore filling was observed. For all materials, loadings above complete pore filling resulted in the recrystallization of PB on the external surface. In vitro drug release measurements showed a rapid dissolution rate at low drug loadings compared to a bimodal release profile of amorphous and crystalline drug at higher drug loadings. PB loaded in mesoporous particle was shown to enhance the antioxidant response to extracellular ROS in the endothelial cell line model, and to intracellular ROS in the microglia cell model. Our results indicate that the antioxidant properties of PB can be significantly improved by using mesoporous silica as a delivery vehicle.
Collapse
|
38
|
Ratajczak K, Lukasiak A, Grel H, Dworakowska B, Jakiela S, Stobiecka M. Monitoring of dynamic ATP level changes by oligomycin-modulated ATP synthase inhibition in SW480 cancer cells using fluorescent "On-Off" switching DNA aptamer. Anal Bioanal Chem 2019; 411:6899-6911. [PMID: 31407049 PMCID: PMC6834760 DOI: 10.1007/s00216-019-02061-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/22/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
Abstract
Adenosine triphosphate (ATP) is the main energy source in cells and an important biomolecule participating in cellular reactions in living organisms. Since the ATP level changes dynamically reflecting the development of a debilitating disease or carcinogenesis, we have focused in this work on monitoring of the oligomycin (OMC)-modulated ATP synthase inhibition using a fluorescent-switching DNA aptamer designed for the detection of ATP (Apt(ATP)), as the model for studies of dynamic ATP level variation. The behavior of the ATP aptamer has been characterized using fluorescence spectroscopy. The Intramolecular fluorescence resonance energy transfer (iFRET) operates in the proposed aptamer from the FAM dye moiety to guanines of the aptamer G-quadruplex when the target ATP is present and binds to the aptamer changing its conformation. The iFRET process enables the detection of ATP down to the limit of detection, LOD = 17 μM, without resorting to any extra chemi-amplification schemes. The selectivity coefficients for relevant interferent triphosphates (UTP, GTP, and CTP) are low for the same concentration as that of ATP. We have demonstrated an efficient transfection of intact cells and OMC-treated SW480 colon cancer cells with Apt(ATP), using microscopic imaging, iFRET measurements, and cell viability testing with MTT method. The applicability of the switching DNA aptamer for the analysis of real samples, obtained by lysis of SW480 cells, was also tested. The proposed Apt(ATP) may be considered as a viable candidate for utilization in measurements of dynamic ATP level modulation in cells in different stages of cancer development and testing of new drugs in pharmacological studies. Graphical abstract ![]()
Collapse
Affiliation(s)
- Katarzyna Ratajczak
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Agnieszka Lukasiak
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Hubert Grel
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Beata Dworakowska
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Slawomir Jakiela
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland.
| | - Magdalena Stobiecka
- Department of Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland.
| |
Collapse
|