151
|
Ferro MP, Heilshorn SC, Owens RM. Materials for blood brain barrier modeling in vitro. MATERIALS SCIENCE & ENGINEERING. R, REPORTS : A REVIEW JOURNAL 2020; 140:100522. [PMID: 33551572 PMCID: PMC7864217 DOI: 10.1016/j.mser.2019.100522] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Brain homeostasis relies on the selective permeability property of the blood brain barrier (BBB). The BBB is formed by a continuous endothelium that regulates exchange between the blood stream and the brain. This physiological barrier also creates a challenge for the treatment of neurological diseases as it prevents most blood circulating drugs from entering into the brain. In vitro cell models aim to reproduce BBB functionality and predict the passage of active compounds through the barrier. In such systems, brain microvascular endothelial cells (BMECs) are cultured in contact with various biomaterial substrates. However, BMEC interactions with these biomaterials and their impact on BBB functions are poorly described in the literature. Here we review the most common materials used to culture BMECs and discuss their potential impact on BBB integrity in vitro. We investigate the biophysical properties of these biomaterials including stiffness, porosity and material degradability. We highlight a range of synthetic and natural materials and present three categories of cell culture dimensions: cell monolayers covering non-degradable materials (2D), cell monolayers covering degradable materials (2.5D) and vascularized systems developing into degradable materials (3D).
Collapse
Affiliation(s)
- Magali P. Ferro
- Department of Bioelectronics, Mines Saint-Étienne, 880 route de Mimet, F-13541, Gardanne, France
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Roisin M. Owens
- Department of Chemical Engineering and Biotechnology, Philippa Fawcett Drive, CB30AS, Cambridge, UK
| |
Collapse
|
152
|
Aoki H, Yamashita M, Hashita T, Iwao T, Matsunaga T. Laminin 221 fragment is suitable for the differentiation of human induced pluripotent stem cells into brain microvascular endothelial-like cells with robust barrier integrity. Fluids Barriers CNS 2020; 17:25. [PMID: 32228708 PMCID: PMC7106710 DOI: 10.1186/s12987-020-00186-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In vitro blood-brain barrier (BBB) models using human induced pluripotent stem (iPS) cell-derived brain microvascular endothelial-like cells (iBMELCs) have been developed to predict the BBB permeability of drug candidates. For the differentiation of iBMELCs, Matrigel, which is a gelatinous protein mixture, is often used as a coating substrate. However, the components of Matrigel can vary among lots, as it is obtained from mouse sarcoma cells with the use of special technics and also contains various basement membranes. Therefore, fully defined substrates as substitutes for Matrigel are needed for a stable supply of iBMELCs with less variation among lots. METHODS iBMELCs were differentiated from human iPS cells on several matrices. The barrier integrity of iBMELCs was evaluated based on transendothelial electrical resistance (TEER) values and permeability of fluorescein isothiocyanate-dextran 4 kDa (FD4) and Lucifer yellow (LY). Characterization of iBMELCs was conducted by RT-qPCR and immunofluorescence analysis. Functions of efflux transporters were defined by intracellular accumulation of the substrates in the wells of multiwell plates. RESULTS iBMELCs differentiated on laminin 221 fragment (LN221F-iBMELCs) had higher TEER values and lower permeability of LY and FD4 as compared with iBMELCs differentiated on Matrigel (Matrigel-iBMELCs). Besides, the gene and protein expression levels of brain microvascular endothelial cells (BMEC)-related markers were similar between LN221F-iBMELCs and Matrigel-iBMELCs. Moreover, both Matrigel- and LN221F-iBMELCs had functions of P-glycoprotein and breast cancer resistance protein, which are essential efflux transporters for barrier functions of the BBB. CONCLUSION The fully defined substrate LN221F presents as an optimal coating matrix for differentiation of iBMELCs. The LN221F-iBMELCs had more robust barrier function for a longer period than Matrigel-iBMELCs with characteristics of BMECs. This finding will contribute the establishment of an iBMELC supply system for pharmacokinetic and pathological models of the BBB.
Collapse
Affiliation(s)
- Hiromasa Aoki
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Misaki Yamashita
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Tadahiro Hashita
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, 467-8603, Japan.
| |
Collapse
|
153
|
Bhalerao A, Sivandzade F, Archie SR, Chowdhury EA, Noorani B, Cucullo L. In vitro modeling of the neurovascular unit: advances in the field. Fluids Barriers CNS 2020; 17:22. [PMID: 32178700 PMCID: PMC7077137 DOI: 10.1186/s12987-020-00183-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
The blood–brain barrier (BBB) is a fundamental component of the central nervous system. Its functional and structural integrity is vital in maintaining the homeostasis of the brain microenvironment. On the other hand, the BBB is also a major hindering obstacle for the delivery of effective therapies to treat disorders of the Central Nervous System (CNS). Over time, various model systems have been established to simulate the complexities of the BBB. The development of realistic in vitro BBB models that accurately mimic the physiological characteristics of the brain microcapillaries in situ is of fundamental importance not only in CNS drug discovery but also in translational research. Successful modeling of the Neurovascular Unit (NVU) would provide an invaluable tool that would aid in dissecting out the pathological factors, mechanisms of action, and corresponding targets prodromal to the onset of CNS disorders. The field of BBB in vitro modeling has seen many fundamental changes in the last few years with the introduction of novel tools and methods to improve existing models and enable new ones. The development of CNS organoids, organ-on-chip, spheroids, 3D printed microfluidics, and other innovative technologies have the potential to advance the field of BBB and NVU modeling. Therefore, in this review, summarize the advances and progress in the design and application of functional in vitro BBB platforms with a focus on rapidly advancing technologies.
Collapse
Affiliation(s)
- Aditya Bhalerao
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Farzane Sivandzade
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Ekram Ahmed Chowdhury
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA
| | - Luca Cucullo
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX, 79106, USA. .,Center for Blood-Brain Barrier Research, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA.
| |
Collapse
|
154
|
Zhang Y, Lu W, Wang Z, Zhang R, Xie Y, Guo S, Jiao L, Hong Y, Di Z, Wang G, Aa J. Reduced Neuronal cAMP in the Nucleus Accumbens Damages Blood-Brain Barrier Integrity and Promotes Stress Vulnerability. Biol Psychiatry 2020; 87:526-537. [PMID: 31812254 DOI: 10.1016/j.biopsych.2019.09.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/09/2019] [Accepted: 09/21/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Studies have suggested that chronic social stress specifically downregulates endothelial tight junction protein expression in the nucleus accumbens (NAc), thus increasing blood-brain barrier (BBB) permeability and promoting depression-like behaviors. However, the molecular mechanism underlying the reduction in tight junction protein, particularly in the NAc, is largely uncharacterized. METHODS We performed comparative metabolomic profiling of the nucleus accumbens, prefrontal cortex, and hippocampus of social defeat-stressed mice to identify the molecular events that mediate BBB breakdown. RESULTS We identified the levels of cyclic adenosine monophosphate (cAMP) that were specifically reduced in the NAc and positively correlated with the degree of social avoidance. Replenishing cAMP in the NAc was sufficient to improve BBB integrity and depression-like behaviors. We further found that cAMP levels were markedly decreased in neurons of the NAc, rather than in endothelial cells, astrocytes, or microglia. RNA-sequencing data showed that adenylate cyclase 5 (Adcy5), an enzyme responsible for the synthesis of cAMP from adenosine triphosphate (ATP), was predominantly expressed in the NAc; it also resided exclusively in neurons. Endogenous modulation of cAMP synthesis in neurons through the knockdown of Adcy5 in the NAc regulated the sensitivity to social stress. Moreover, deficient neuronal cAMP production in the NAc decreased the expression of reelin, while supplementary injection of exogenous reelin into the NAc promoted BBB integrity and ameliorated depression-like behaviors. CONCLUSIONS Chronic social stress diminished cAMP synthesis in neurons, thus damaging BBB integrity in the NAc and promoting stress vulnerability. These results characterize neuron-produced cAMP in the NAc as a biological mechanism of neurovascular pathology in social stress.
Collapse
Affiliation(s)
- Yue Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wuhuan Lu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zibin Wang
- Analytical and Testing Center, Nanjing Medical University, Nanjing, China
| | - Ran Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yuan Xie
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Suhan Guo
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Li Jiao
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu Hong
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zizhen Di
- Liaoning Provincial Academy of Traditional Chinese Medicine, Shenyang, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| | - Jiye Aa
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
155
|
Li S, Wang Y, Jiang D, Ni D, Kutyreff CJ, Barnhart TE, Engle JW, Cai W. Spatiotemporal Distribution of Agrin after Intrathecal Injection and Its Protective Role in Cerebral Ischemia/Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902600. [PMID: 32076591 PMCID: PMC7029627 DOI: 10.1002/advs.201902600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/18/2019] [Indexed: 05/30/2023]
Abstract
Intrathecal injection, drugs transporting along perivascular spaces, represents an important route for maintaining blood-brain barrier (BBB) integrity after cerebral ischemia/reperfusion (I/R) injury. However, after being directly injected into cerebrospinal fluid (CSF), the temporal and spatial changes in the distribution of therapeutic protein drugs have remained unknown. Here, with positron emission tomography (PET) imaging, the uptake of 89Zr-agrin is noninvasively and dynamically monitored. These data demonstrate the time-activity curve of drugs in the brain subregions and their spatial distribution in different organs after intrathecal administration. Furthermore, agrin treatment effectively inhibits BBB disruption by reducing the loss of tight-junctional proteins. Importantly, the infarct volume is reduced; the number of apoptotic neurons is decreased; and neurological function is improved in mouse I/R injury models. Thus, intrathecal injection of agrin provides the basis for a new strategy to research and develop protein drugs for reducing the aggravation of I/R injury.
Collapse
Affiliation(s)
- Shiyong Li
- Department of RehabilitationSecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Ye Wang
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
| | - Dawei Jiang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Dalong Ni
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Christopher J. Kutyreff
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Todd E. Barnhart
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Jonathan W. Engle
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| |
Collapse
|
156
|
The Challenge of Modulating Heparan Sulfate Turnover by Multitarget Heparin Derivatives. Molecules 2020; 25:molecules25020390. [PMID: 31963505 PMCID: PMC7024324 DOI: 10.3390/molecules25020390] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
This review comes as a part of the special issue "Emerging frontiers in GAGs and mimetics". Our interest is in the manipulation of heparan sulfate (HS) turnover by employing HS mimetics/heparin derivatives that exert pleiotropic effects and are interesting for interfering at multiple levels with pathways in which HS is implicated. Due to the important role of heparanase in HS post-biosynthetic modification and catabolism, we focus on the possibility to target heparanase, at both extracellular and intracellular levels, a strategy that can be applied to many conditions, from inflammation to cancer and neurodegeneration.
Collapse
|
157
|
Guo Q, Yang S, Yang D, Zhang N, Li X, Chen T, Chen J, Li G, Yin L, Wu Q. Differential mRNA expression combined with network pharmacology reveals network effects of Liangxue Tongyu Prescription for acute intracerebral hemorrhagic rats. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112231. [PMID: 31520671 DOI: 10.1016/j.jep.2019.112231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liangxue Tongyu Prescription (LTP) is a traditional Chinese medicine formula composed of 8 crude drugs that is widely used to treat acute intracerebral hemorrhage (AICH). AIM OF THE STUDY To verify the efficacy of LTP on the survival time in the treatment of acute intracerebral hemorrhagic rats (AICHs), and to elucidate its network pharmacodynamic mechanism of multi-component, multi-target, and multi-signaling pathways. MATERIALS AND METHODS Survival analysis was used to evaluate the survival time of AICH rats induced by different doses of collagenase and the efficacy of three doses of LTP in the treatment of AICH rats. The Kaplan-Meier curves for survival time were produced and compared with the Log-rank test and Wilcoxon (Gehan) χ2. Differential mRNA-seq combined with network pharmacology was used to disclose the network effect mechanism of LTP on AICH, and the obtained differential genes were mapped into the predictive empirical compound-target network model (ECT network model) and the empirical compound-target-pathogenesis (disease) network model (ECTP network model). RESULTS The median survival time of four different doses of LTP-treated groups (0.00 g/kg, 5.78 g/kg, 11.55 g/kg, 23.10 g/kg) for adult AICH rats by 0.18 U collagenase was 14 h, 37 h, 150 h, and 51 h respectively, and the 7-day survival rates were 33.3%, 41.7%, 50.0%, and 38.5%, of which the medium-dose group (MD) had a longer survival time and higher survival rate. Through further validation experiments, the MD group had a better efficacy trend with a median survival time of 168 h vs 23 h in the model control group (MC) (Wilcoxon Gehan Test, χ2 = 3.478, P = 0.062). The transcriptomic analysis of mRNA showed that 583 significant differential genes were found between the MC and MD group and 7 key therapeutic targets regulated by 29 compounds in LTP on AICH were screened out by VCT and VCTP network model. These targets were involved in 5 regulatory models or pathways. CONCLUSION Our study confirmed the exact efficacy of the LTP in the treatment of AICH and revealed the potential pharmacodynamic components and mode of action of the LTP on AICH. Using differential transcriptome of mRNA combined with network pharmacology, we screened out 29 chemical compounds as the potential effective ingredients of LTP which acted on 7 targets of AICH involving 5 pathological pathways, mainly including repairing the brain function defect, improving neural function, protecting blood-brain barrier from damage, reducing inflammatory factors, and inhibiting apoptosis. The present study not only provides a new explanation for the 'multi-component, multi-target, multi-pathway' effects of the LTP on AICH but also screened out some major compounds of LTP and their potential targets which will facilitate the development of new drugs for AICH.
Collapse
Affiliation(s)
- Qingqing Guo
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China
| | - Shijin Yang
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China
| | - Dongqing Yang
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China
| | - Ning Zhang
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China
| | - Xun Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Nanjing, PR China
| | - Tianli Chen
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China
| | - Jiayan Chen
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China
| | - Guochun Li
- Department of Public Health, Nanjing University of Chinese Medicine, 210023, Nanjing, PR China.
| | - Lian Yin
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Nanjing, PR China.
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, PR China.
| |
Collapse
|
158
|
Li Y, Wu P, Bihl JC, Shi H. Underlying Mechanisms and Potential Therapeutic Molecular Targets in Blood-Brain Barrier Disruption after Subarachnoid Hemorrhage. Curr Neuropharmacol 2020; 18:1168-1179. [PMID: 31903882 PMCID: PMC7770641 DOI: 10.2174/1570159x18666200106154203] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/18/2019] [Accepted: 01/04/2020] [Indexed: 01/01/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a subtype of hemorrhagic stroke with significant morbidity and mortality. Aneurysmal bleeding causes elevated intracranial pressure, decreased cerebral blood flow, global cerebral ischemia, brain edema, blood component extravasation, and accumulation of breakdown products. These post-SAH injuries can disrupt the integrity and function of the blood-brain barrier (BBB), and brain tissues are directly exposed to the neurotoxic blood contents and immune cells, which leads to secondary brain injuries including inflammation and oxidative stress, and other cascades. Though the exact mechanisms are not fully clarified, multiple interconnected and/or independent signaling pathways have been reported to be involved in BBB disruption after SAH. In addition, alleviation of BBB disruption through various pathways or chemicals has a neuroprotective effect on SAH. Hence, BBB permeability plays an important role in the pathological course and outcomes of SAH. This review discusses the recent understandings of the underlying mechanisms and potential therapeutic targets in BBB disruption after SAH, emphasizing the dysfunction of tight junctions and endothelial cells in the development of BBB disruption. The emerging molecular targets, including toll-like receptor 4, netrin-1, lipocalin-2, tropomyosin-related kinase receptor B, and receptor tyrosine kinase ErbB4, are also summarized in detail. Finally, we discussed the emerging treatments for BBB disruption after SAH and put forward our perspectives on future research.
Collapse
Affiliation(s)
| | | | - Ji C. Bihl
- Address correspondence to these authors at the Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, 45435, USA; Tel: 011-01-9377755243; Fax: 011-01-9377757221; E-mail: and Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China; Tel: +86-15545107889; E-mail:
| | - Huaizhang Shi
- Address correspondence to these authors at the Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, 45435, USA; Tel: 011-01-9377755243; Fax: 011-01-9377757221; E-mail: and Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China; Tel: +86-15545107889; E-mail:
| |
Collapse
|
159
|
Yamazaki Y, Shinohara M, Yamazaki A, Ren Y, Asmann YW, Kanekiyo T, Bu G. ApoE (Apolipoprotein E) in Brain Pericytes Regulates Endothelial Function in an Isoform-Dependent Manner by Modulating Basement Membrane Components. Arterioscler Thromb Vasc Biol 2020; 40:128-144. [PMID: 31665905 PMCID: PMC7007705 DOI: 10.1161/atvbaha.119.313169] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/17/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The ε4 allele of the APOE gene (APOE4) is the strongest genetic risk factor for Alzheimer disease when compared with the common ε3 allele. Although there has been significant progress in understanding how apoE4 (apolipoprotein E4) drives amyloid pathology, its effects on amyloid-independent pathways, in particular cerebrovascular integrity and function, are less clear. Approach and Results: Here, we show that brain pericytes, the mural cells of the capillary walls, differentially modulate endothelial cell phenotype in an apoE isoform-dependent manner. Extracellular matrix protein induction, tube-like structure formation, and barrier formation were lower with endothelial cells cocultured with pericytes isolated from apoE4-targeted replacement (TR) mice compared with those from apoE3-TR mice. Importantly, aged apoE4-targeted replacement mice had decreased extracellular matrix protein expression and increased plasma protein leakages compared with apoE3-TR mice. CONCLUSIONS ApoE4 impairs pericyte-mediated basement membrane formation, potentially contributing to the cerebrovascular effects of apoE4.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | | | - Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| | - Yingxue Ren
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224
| | - Yan W. Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224
| | | | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224
| |
Collapse
|
160
|
Effect of chronic methylphenidate treatment on hippocampal neurovascular unit and memory performance in late adolescent rats. Eur Neuropsychopharmacol 2019; 29:195-210. [PMID: 30554860 DOI: 10.1016/j.euroneuro.2018.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/13/2018] [Accepted: 12/01/2018] [Indexed: 12/21/2022]
Abstract
Methylphenidate (MPH) is the classic treatment for attention deficit hyperactivity disorder (ADHD) among children and adults. Despite its beneficial effects, non-medical use of MPH is nowadays a problem with high impact on society. Thus, our goal was to uncover the neurovascular and cognitive effects of MPH chronic use during a critical period of development in control conditions. For that, male Wistar Kyoto rats were treated with MPH (1.5 or 5 mg/kg/day at weekdays, per os) from P28 to P55. We concluded that the higher dose of MPH caused hippocampal blood-brain barrier (BBB) hyperpermeability by vesicular transport (transcytosis) concomitantly with the presence of peripheral immune cells in the brain parenchyma. These observations were confirmed by in vitro studies, in which the knockdown of caveolin-1 in human brain endothelial cells prevented the increased permeability and leukocytes transmigration triggered by MPH (100 µM, 24 h). Furthermore, MPH led to astrocytic atrophy and to a decrease in the levels of several synaptic proteins and impairment of AKT/CREB signaling, together with working memory deficit assessed in the Y-maze test. On the contrary, we verified that the lower dose of MPH (1.5 mg/kg/day) increased astrocytic processes and upregulated several neuronal proteins as well as signaling pathways involved in synaptic plasticity culminating in working memory improvement. In conclusion, the present study reveals that a lower dose of MPH in normal rats improves memory performance being associated with the modulation of astrocytic morphology and synaptic machinery. However, a higher dose of MPH leads to BBB dysfunction and memory impairment.
Collapse
|
161
|
Elastin-Derived Peptide VGVAPG Affects Production and Secretion of Testosterone in Mouse Astrocyte In Vitro. Neurochem Res 2019; 45:385-394. [PMID: 31776971 DOI: 10.1007/s11064-019-02920-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/18/2019] [Accepted: 11/25/2019] [Indexed: 12/18/2022]
Abstract
Astrocytes play many distinct roles in the nervous system providing structural support for neurons and maintaining blood-brain barrier integrity. Steroid hormones exhibit a broad spectrum of actions in the central and peripheral nervous system, acting as trophic factors affecting cell differentiation and synaptic plasticity. In steroidogenesis, astrocytes play a key role by producing cholesterol, progesterone (P4), testosterone (T), and estradiol (E2). Currently there are only few studies which show that the Gly-Val-Ala-Pro-Gly (VGVAPG) peptide may affect the metabolism of astrocytes. Therefore, due to the role of neurosteroids, it is necessary to determine whether VGVAPG affects the level of E2, P4, and T in astrocytes. Primary mouse astrocytes were maintained in DMEM/F12 without phenol red, and supplemented with 10% charcoal/dextran-treated fetal bovine serum. Cells were exposed to 10 nM and 1 µM VGVAPG peptide and co-treated with cSrc kinase inhibitor I. After cell stimulation, we measured the Ki67 protein level and the production and secretion of P4, T, and E2. Our report presents the novel finding that the VGVAPG peptide affects the production and secretion of neurosteroids in astrocytes in vitro. The VGVAPG peptide increases the production of P4; however, at the same time, it decreases the secretion of P4 by astrocytes. On the other hand, it stimulates the production and secretion of T. Interestingly, the production of E2 did not change in any studied time interval. The expression of Ki67 protein increased after 48 h of exposition to the VGVAPG peptide. The cSrc kinase inhibitor I prevented most of the effects of VGVAPG peptide. Therefore, we postulate that T and cSrc kinase may be responsible for increasing astrocyte proliferation.
Collapse
|
162
|
Balikov DA, Neal EH, Lippmann ES. Organotypic Neurovascular Models: Past Results and Future Directions. Trends Mol Med 2019; 26:273-284. [PMID: 31699496 DOI: 10.1016/j.molmed.2019.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
The high failure rates of clinical trials in neurodegeneration, perhaps most apparent in recent high-profile failures of potential Alzheimer's disease therapies, have partially motivated the development of improved human cell-based models to bridge the gap between well-plate assays and preclinical efficacy studies in mice. Recently, cerebral organoids derived from stem cells have gained significant traction as 3D models of central nervous system (CNS) regions. Although this technology is promising, several limitations still exist; most notably, improper structural organization of neural cells and a lack of functional glia and vasculature. Here, we provide an overview of the cerebral organoid field and speculate how engineering strategies, including biomaterial fabrication and templating, might be used to overcome existing challenges.
Collapse
Affiliation(s)
- Daniel A Balikov
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Emma H Neal
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Chemical and Physical Biology Program, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
163
|
Nakamura K, Ikeuchi T, Nara K, Rhodes CS, Zhang P, Chiba Y, Kazuno S, Miura Y, Ago T, Arikawa-Hirasawa E, Mukouyama YS, Yamada Y. Perlecan regulates pericyte dynamics in the maintenance and repair of the blood-brain barrier. J Cell Biol 2019; 218:3506-3525. [PMID: 31541017 PMCID: PMC6781430 DOI: 10.1083/jcb.201807178] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/10/2019] [Accepted: 07/31/2019] [Indexed: 01/24/2023] Open
Abstract
Ischemic stroke causes blood-brain barrier (BBB) breakdown due to significant damage to the integrity of BBB components. Recent studies have highlighted the importance of pericytes in the repair process of BBB functions triggered by PDGFRβ up-regulation. Here, we show that perlecan, a major heparan sulfate proteoglycan of basement membranes, aids in BBB maintenance and repair through pericyte interactions. Using a transient middle cerebral artery occlusion model, we found larger infarct volumes and more BBB leakage in conditional perlecan (Hspg2)-deficient (Hspg2 - / - -TG) mice than in control mice. Control mice showed increased numbers of pericytes in the ischemic lesion, whereas Hspg2 - / - -TG mice did not. At the mechanistic level, pericytes attached to recombinant perlecan C-terminal domain V (perlecan DV, endorepellin). Perlecan DV enhanced the PDGF-BB-induced phosphorylation of PDGFRβ, SHP-2, and FAK partially through integrin α5β1 and promoted pericyte migration. Perlecan therefore appears to regulate pericyte recruitment through the cooperative functioning of PDGFRβ and integrin α5β1 to support BBB maintenance and repair following ischemic stroke.
Collapse
Affiliation(s)
- Kuniyuki Nakamura
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD .,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoko Ikeuchi
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Kazuki Nara
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD.,Tohoku University School of Medicine, Sendai, Japan
| | - Craig S Rhodes
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Peipei Zhang
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Yuta Chiba
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eri Arikawa-Hirasawa
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Yoshihiko Yamada
- Molecular Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| |
Collapse
|
164
|
Nowak M, Helgeson ME, Mitragotri S. Delivery of Nanoparticles and Macromolecules across the Blood–Brain Barrier. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900073] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Maksymilian Nowak
- School of Engineering and Applied Sciences Harvard University 29 Oxford St. Cambridge MA 02318 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University 3 Blackfan Circle Boston MA 02115 USA
| | - Matthew E. Helgeson
- Department of Chemical Engineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences Harvard University 29 Oxford St. Cambridge MA 02318 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University 3 Blackfan Circle Boston MA 02115 USA
| |
Collapse
|
165
|
Reed MJ, Damodarasamy M, Banks WA. The extracellular matrix of the blood-brain barrier: structural and functional roles in health, aging, and Alzheimer's disease. Tissue Barriers 2019; 7:1651157. [PMID: 31505997 DOI: 10.1080/21688370.2019.1651157] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
There is increasing interest in defining the location, content, and role of extracellular matrix (ECM) components in brain structure and function during development, aging, injury, and neurodegeneration. Studies in vivo confirm brain ECM has a dynamic composition with constitutive and induced alterations that impact subsequent cell-cell and cell-matrix interactions. Moreover, it is clear that for any given ECM component, the brain region, and cell type within that location, determines the direction, magnitude, and composition of those changes. This review will examine the ECM at the neurovascular unit (NVU) and the blood-brain barrier (BBB) within the NVU. The discussion will begin at the glycocalyx ECM on the luminal surface of the vasculature, and progress to the abluminal side with a focus on changes in basement membrane ECM during aging and neurodegeneration.
Collapse
Affiliation(s)
- May J Reed
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - Mamatha Damodarasamy
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA
| | - William A Banks
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington, Seattle, WA, USA.,VA Puget Sound Health Care System, Geriatric Research Education and Clinical Center, Seattle, WA, USA
| |
Collapse
|
166
|
Praça C, Rosa SC, Sevin E, Cecchelli R, Dehouck MP, Ferreira LS. Derivation of Brain Capillary-like Endothelial Cells from Human Pluripotent Stem Cell-Derived Endothelial Progenitor Cells. Stem Cell Reports 2019; 13:599-611. [PMID: 31495714 PMCID: PMC6829749 DOI: 10.1016/j.stemcr.2019.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
The derivation of human brain capillary endothelial cells is of utmost importance for drug discovery programs focusing on diseases of the central nervous system. Here, we describe a two-step differentiation protocol to derive brain capillary-like endothelial cells from human pluripotent stem cells. The cells were initially differentiated into endothelial progenitor cells followed by specification into a brain capillary-like endothelial cell phenotype using a protocol that combined the induction, in a time-dependent manner, of VEGF, Wnt3a, and retinoic acid signaling pathways and the use of fibronectin as the extracellular matrix. The brain capillary-like endothelial cells displayed a permeability to lucifer yellow of 1 × 10−3 cm/min, a transendothelial electrical resistance value of 60 Ω cm2 and were able to generate a continuous monolayer of cells expressing ZO-1 and CLAUDIN-5 but moderate expression of P-glycoprotein. Further maturation of these cells required coculture with pericytes. The study presented here opens a new approach for the study of soluble and non-soluble factors in the specification of endothelial progenitor cells into brain capillary-like endothelial cells. Derivation of BCLECs from iPSCs in chemically defined medium in two steps Specification of EPCs into BCLECs requires the activation of VEGF, Wnt3a, and RA Fibronectin seems a sufficient substrate for the specification of EPCs into BCLECs
Collapse
Affiliation(s)
- Catarina Praça
- Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Susana C Rosa
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Emmanuel Sevin
- Faculté des Sciences Jean Perrin, Université d'Artois, Lens, France
| | - Romeo Cecchelli
- Faculté des Sciences Jean Perrin, Université d'Artois, Lens, France
| | | | - Lino S Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
167
|
Nakagomi T, Takagi T, Beppu M, Yoshimura S, Matsuyama T. Neural regeneration by regionally induced stem cells within post-stroke brains: Novel therapy perspectives for stroke patients. World J Stem Cells 2019; 11:452-463. [PMID: 31523366 PMCID: PMC6716084 DOI: 10.4252/wjsc.v11.i8.452] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/04/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a critical disease which causes serious neurological functional loss such as paresis. Hope for novel therapies is based on the increasing evidence of the presence of stem cell populations in the central nervous system (CNS) and the development of stem-cell-based therapies for stroke patients. Although mesenchymal stem cells (MSCs) represented initially a promising cell source, only a few transplanted MSCs were present near the injured areas of the CNS. Thus, regional stem cells that are present and/or induced in the CNS may be ideal when considering a treatment following ischemic stroke. In this context, we have recently showed that injury/ischemia-induced neural stem/progenitor cells (iNSPCs) and injury/ischemia-induced multipotent stem cells (iSCs) are present within post-stroke human brains and post-stroke mouse brains. This indicates that iNSPCs/iSCs could be developed for clinical applications treating patients with stroke. The present study introduces the traits of mouse and human iNSPCs, with a focus on the future perspective for CNS regenerative therapies using novel iNSPCs/iSCs.
Collapse
Affiliation(s)
- Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Toshinori Takagi
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Mikiya Beppu
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshimura
- Department of Neurosurgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Tomohiro Matsuyama
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
168
|
Glycation-induced modification of tissue-specific ECM proteins: A pathophysiological mechanism in degenerative diseases. Biochim Biophys Acta Gen Subj 2019; 1863:129411. [PMID: 31400438 DOI: 10.1016/j.bbagen.2019.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Glycation driven generation of advanced glycation end products (AGEs) and their patho-physiological role in human degenerative diseases has remained one of the thrust areas in the mainstream of disease biology. Glycation of extracellular matrix (ECM) proteins have deleterious effect on the mechanical and functional properties of tissues. Owing to the adverse pathophysiological concerns of glycation, there is a need to decipher the underlying mechanisms. SCOPE OF REVIEW AGE-modified ECM proteins affect the cell in the vicinity by altering protein structure-function, matrix-matrix or matrix-cell interaction and by activating signalling pathway through receptor for AGE. This review is intended for addressing the AGE-induced modification of tissue-specific ECM proteins and its implication in the pathogenesis of various organ-specific human ailments. MAJOR CONCLUSIONS The glycation affects the canonical cell behaviour due to alteration in the interaction of glycated ECM with receptors like integrins and discodin domain, and the signalling cues generated subsequently affect the downstream signalling pathways. Consequently, the variation of structural and functional properties of tissues due to matrix glycation helps in the initiation or progression of the disease condition. GENERAL SIGNIFICANCE This review offers comprehensive knowledge about the remodelling of glycation induced ECM and tissue-specific pathological concerns. As glycation of ECM affects the normal tissues and cell behaviour, the scientific discourse may also provide cues for developing candidate drugs that may help in attenuating the adverse effects of AGEs and perhaps open a research window of tailoring novel strategies for the management of glycation induced human degenerative diseases.
Collapse
|
169
|
Ferreira L. What human blood-brain barrier models can tell us about BBB function and drug discovery? Expert Opin Drug Discov 2019; 14:1113-1123. [DOI: 10.1080/17460441.2019.1646722] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
170
|
de la Vega L, Lee C, Sharma R, Amereh M, Willerth SM. 3D bioprinting models of neural tissues: The current state of the field and future directions. Brain Res Bull 2019; 150:240-249. [DOI: 10.1016/j.brainresbull.2019.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
|
171
|
Nizari S, Carare RO, Romero IA, Hawkes CA. 3D Reconstruction of the Neurovascular Unit Reveals Differential Loss of Cholinergic Innervation in the Cortex and Hippocampus of the Adult Mouse Brain. Front Aging Neurosci 2019; 11:172. [PMID: 31333445 PMCID: PMC6620643 DOI: 10.3389/fnagi.2019.00172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/20/2019] [Indexed: 01/02/2023] Open
Abstract
Increasing evidence supports a role for cerebrovasculature dysfunction in the etiology of Alzheimer’s disease (AD). Blood vessels in the brain are composed of a collection of cells and acellular material that comprise the neurovascular unit (NVU). The NVU in the hippocampus and cortex receives innervation from cholinergic neurons that originate in the basal forebrain. Death of these neurons and their nerve fibers is an early feature of AD. However, the effect of the loss of cholinergic innervation on the NVU is not well characterized. The purpose of this study was to evaluate the effect of the loss of cholinergic innervation of components of the NVU at capillaries, arteries and veins in the hippocampus and cortex. Adult male C57BL/6 mice received an intracerebroventricular injection of the immunotoxin p75NTR mu-saporin to induce the loss of cholinergic neurons. Quadruple labeling immunohistochemistry and 3D reconstruction were carried out to characterize specific points of contact between cholinergic fibers and collagen IV, smooth muscle cells and astrocyte endfeet. Innate differences were observed between vessels of the hippocampus and cortex of control mice, including a greater amount of cholinergic contact with perivascular astrocytes in hippocampal capillaries and a thicker basement membrane in hippocampal veins. Saporin treatment induced a loss of cholinergic innervation at the arterial basement membrane and smooth muscle cells of both the hippocampus and the cortex. In the cortex, there was an additional loss of innervation at the astrocytic endfeet. The current results suggest that cortical arteries are more strongly affected by cholinergic denervation than arteries in the hippocampus. This regional variation may have implications for the etiology of the vascular pathology that develops in AD.
Collapse
Affiliation(s)
- Shereen Nizari
- School of Life, Health and Chemical Science, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Roxana O Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ignacio A Romero
- School of Life, Health and Chemical Science, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| | - Cheryl A Hawkes
- School of Life, Health and Chemical Science, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes, United Kingdom
| |
Collapse
|
172
|
Park TE, Mustafaoglu N, Herland A, Hasselkus R, Mannix R, FitzGerald EA, Prantil-Baun R, Watters A, Henry O, Benz M, Sanchez H, McCrea HJ, Goumnerova LC, Song HW, Palecek SP, Shusta E, Ingber DE. Hypoxia-enhanced Blood-Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat Commun 2019; 10:2621. [PMID: 31197168 DOI: 10.1101/482463v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/16/2019] [Indexed: 05/21/2023] Open
Abstract
The high selectivity of the human blood-brain barrier (BBB) restricts delivery of many pharmaceuticals and therapeutic antibodies to the central nervous system. Here, we describe an in vitro microfluidic organ-on-a-chip BBB model lined by induced pluripotent stem cell-derived human brain microvascular endothelium interfaced with primary human brain astrocytes and pericytes that recapitulates the high level of barrier function of the in vivo human BBB for at least one week in culture. The endothelium expresses high levels of tight junction proteins and functional efflux pumps, and it displays selective transcytosis of peptides and antibodies previously observed in vivo. Increased barrier functionality was accomplished using a developmentally-inspired induction protocol that includes a period of differentiation under hypoxic conditions. This enhanced BBB Chip may therefore represent a new in vitro tool for development and validation of delivery systems that transport drugs and therapeutic antibodies across the human BBB.
Collapse
Affiliation(s)
- Tae-Eun Park
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Nur Mustafaoglu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ryan Hasselkus
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Edward A FitzGerald
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Alexander Watters
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Olivier Henry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Maximilian Benz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Henry Sanchez
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Heather J McCrea
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Hannah W Song
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Eric Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
173
|
Park TE, Mustafaoglu N, Herland A, Hasselkus R, Mannix R, FitzGerald EA, Prantil-Baun R, Watters A, Henry O, Benz M, Sanchez H, McCrea HJ, Goumnerova LC, Song HW, Palecek SP, Shusta E, Ingber DE. Hypoxia-enhanced Blood-Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat Commun 2019; 10:2621. [PMID: 31197168 PMCID: PMC6565686 DOI: 10.1038/s41467-019-10588-0] [Citation(s) in RCA: 348] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/16/2019] [Indexed: 12/20/2022] Open
Abstract
The high selectivity of the human blood-brain barrier (BBB) restricts delivery of many pharmaceuticals and therapeutic antibodies to the central nervous system. Here, we describe an in vitro microfluidic organ-on-a-chip BBB model lined by induced pluripotent stem cell-derived human brain microvascular endothelium interfaced with primary human brain astrocytes and pericytes that recapitulates the high level of barrier function of the in vivo human BBB for at least one week in culture. The endothelium expresses high levels of tight junction proteins and functional efflux pumps, and it displays selective transcytosis of peptides and antibodies previously observed in vivo. Increased barrier functionality was accomplished using a developmentally-inspired induction protocol that includes a period of differentiation under hypoxic conditions. This enhanced BBB Chip may therefore represent a new in vitro tool for development and validation of delivery systems that transport drugs and therapeutic antibodies across the human BBB.
Collapse
Affiliation(s)
- Tae-Eun Park
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Nur Mustafaoglu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ryan Hasselkus
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Edward A FitzGerald
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Alexander Watters
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Olivier Henry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Maximilian Benz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Henry Sanchez
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Heather J McCrea
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Hannah W Song
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Eric Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
174
|
Oddo A, Peng B, Tong Z, Wei Y, Tong WY, Thissen H, Voelcker NH. Advances in Microfluidic Blood-Brain Barrier (BBB) Models. Trends Biotechnol 2019; 37:1295-1314. [PMID: 31130308 DOI: 10.1016/j.tibtech.2019.04.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 12/21/2022]
Abstract
Therapeutic options for neurological disorders currently remain limited. The intrinsic complexity of the brain architecture prevents potential therapeutics from reaching their cerebral target, thus limiting their efficacy. Recent advances in microfluidic technology and organ-on-chip systems have enabled the development of a new generation of in vitro platforms that can recapitulate complex in vivo microenvironments and physiological responses. In this context, microfluidic-based in vitro models of the blood-brain barrier (BBB) are of particular interest as they provide an innovative approach for conducting research related to the brain, including modeling of neurodegenerative diseases and high-throughput drug screening. Here, we present the most recent advances in BBB-on-chip devices and examine validation steps that will strengthen their future applications.
Collapse
Affiliation(s)
- Arianna Oddo
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Bo Peng
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia.
| | - Ziqiu Tong
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Yingkai Wei
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Wing Yin Tong
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Helmut Thissen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Nicolas Hans Voelcker
- Drug Delivery, Disposition, and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia; Department of Materials Science and Engineering, Monash University, Clayton, Victoria 3168, Australia.
| |
Collapse
|
175
|
Jafari B, Pourseif MM, Barar J, Rafi MA, Omidi Y. Peptide-mediated drug delivery across the blood-brain barrier for targeting brain tumors. Expert Opin Drug Deliv 2019; 16:583-605. [DOI: 10.1080/17425247.2019.1614911] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Behzad Jafari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia,
Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Mohammad A. Rafi
- Department of Neurology, College of Medicine, Thomas Jefferson University, Philadelphia,
PA, USA
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| |
Collapse
|
176
|
Lee TH, Hsieh ST, Chiang HY. Fibronectin inhibitor pUR4 attenuates tumor necrosis factor α-induced endothelial hyperpermeability by modulating β1 integrin activation. J Biomed Sci 2019; 26:37. [PMID: 31096970 PMCID: PMC6521375 DOI: 10.1186/s12929-019-0529-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 05/05/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The blood-spinal cord barrier (BSCB) is composed of a monolayer of endothelium linked with tight junctions and extracellular matrix (ECM)-rich basement membranes and is surrounded by astrocyte foot processes. Endothelial permeability is regulated by interaction between endothelial cells and ECM proteins. Fibronectin (FN) is a principal ECM component of microvessels. Excessive FN deposition disrupts cell-cell adhesion in fibroblasts through β1 integrin ligation. To determine whether excessive FN deposition contributes to the disruption of endothelial integrity, we used an in vitro model of the endothelial monolayer to investigate whether the FN inhibitor pUR4 prevents FN deposition into the subendothelial matrix and attenuates endothelial leakage. METHODS To correlate the effects of excessive FN accumulation in microvessels on BSCB disruption, spinal nerve ligation-which induces BSCB leakage-was applied, and FN expression in the spinal cord was evaluated through immunohistochemistry and immunoblotting. To elucidate the effects by which pUR4 modulates endothelial permeability, brain-derived endothelial (bEND.3) cells treated with tumor necrosis factor (TNF)-α were used to mimic a leaky BSCB. A bEND.3 monolayer was preincubated with pUR4 before TNF-α treatment. The transendothelial electrical resistance (TEER) measurement and transendothelial permeability assay were applied to assess the endothelial integrity of the bEND.3 monolayer. Immunofluorescence analysis and immunoblotting were performed to evaluate the inhibitory effects of pUR4 on TNF-α-induced FN deposition. To determine the mechanisms underlying pUR4-mediated endothelial permeability, cell morphology, stress fiber formation, myosin light chain (MLC) phosphorylation, and β1 integrin-mediated signaling were evaluated through immunofluorescence analysis and immunoblotting. RESULTS Excessive FN was accumulated in the microvessels of the spinal cord after spinal nerve ligation; moreover, pUR4 inhibited TNF-α-induced FN deposition in the bEND.3 monolayer and maintained intact TEER and endothelial permeability. Furthermore, pUR4 reduced cell morphology alteration, actin stress fiber formation, and MLC phosphorylation, thereby attenuating paracellular gap formation. Moreover, pUR4 reduced β1 integrin activation and downstream signaling. CONCLUSIONS pUR4 reduces TNF-α-induced β1 integrin activation by depleting ECM FN, leading to a decrease in endothelial hyperpermeability and maintenance of monolayer integrity. These findings suggest therapeutic benefits of pUR4 in pathological vascular leakage treatment.
Collapse
Affiliation(s)
- Ting-Hein Lee
- Department of Anatomy, College of Medicine, Chang Gung University, 259 Wenhua 1st Rd., Guishan Dist, Taoyuan City, 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hou-Yu Chiang
- Department of Anatomy, College of Medicine, Chang Gung University, 259 Wenhua 1st Rd., Guishan Dist, Taoyuan City, 33302, Taiwan. .,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan. .,Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
177
|
Chen-Roetling J, Cao Y, Peng D, Regan RF. Rapid loss of perihematomal cell viability in the collagenase intracerebral hemorrhage model. Brain Res 2019; 1711:91-96. [PMID: 30639124 PMCID: PMC6519080 DOI: 10.1016/j.brainres.2019.01.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 01/24/2023]
Abstract
The effective time window of any therapeutic in an experimental stroke model is limited by the rate of injury progression. Intracerebral hemorrhage in rodents is commonly induced by striatal injection of either autologous blood or bacterial collagenase, which digests local blood vessels. During time window studies of the heme oxygenase-1 inducer hemin, which is protective when administered within 1-3 h in both models, the rate of perihematomal injury was directly compared after striatal blood or collagenase injection. Surprisingly, about 80% of the loss of perihematomal cell viability as measured by MTT reduction assay occurred within 6 h of collagenase injection. In contrast, significant viability loss was not observed at this time point after autologous blood injection, but rather it progressed over the subsequent four days to a level similar to that produced by collagenase. Consistent with these observations, systemic hemin therapy reduced blood-brain barrier disruption and perihematomal cell injury when initiated at 6 h after striatal injection of blood but not collagenase. These results indicate that the rate of early cell injury differs markedly in the collagenase and blood injection ICH models, which may contribute to inconsistent results in time window studies. The blood injection model may be more appropriate for prolonged time window studies of a neuroprotective agent.
Collapse
Affiliation(s)
| | - Yang Cao
- Department of Emergency Medicine, Thomas Jefferson University, USA
| | - Denggao Peng
- Department of Emergency Medicine, Thomas Jefferson University, USA
| | - Raymond F Regan
- Department of Emergency Medicine, Thomas Jefferson University, USA.
| |
Collapse
|
178
|
Jensen LD, Hot B, Ramsköld D, Germano RFV, Yokota C, Giatrellis S, Lauschke VM, Hubmacher D, Li MX, Hupe M, Arnold TD, Sandberg R, Frisén J, Trusohamn M, Martowicz A, Wisniewska-Kruk J, Nyqvist D, Adams RH, Apte SS, Vanhollebeke B, Stenman JM, Kele J. Disruption of the Extracellular Matrix Progressively Impairs Central Nervous System Vascular Maturation Downstream of β-Catenin Signaling. Arterioscler Thromb Vasc Biol 2019; 39:1432-1447. [PMID: 31242033 PMCID: PMC6597191 DOI: 10.1161/atvbaha.119.312388] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— The Wnt/β-catenin pathway orchestrates development of the blood-brain barrier, but the downstream mechanisms involved at different developmental windows and in different central nervous system (CNS) tissues have remained elusive. Approach and Results— Here, we create a new mouse model allowing spatiotemporal investigations of Wnt/β-catenin signaling by induced overexpression of Axin1, an inhibitor of β-catenin signaling, specifically in endothelial cells (Axin1iEC−OE). AOE (Axin1 overexpression) in Axin1iEC−OE mice at stages following the initial vascular invasion of the CNS did not impair angiogenesis but led to premature vascular regression followed by progressive dilation and inhibition of vascular maturation resulting in forebrain-specific hemorrhage 4 days post-AOE. Analysis of the temporal Wnt/β-catenin driven CNS vascular development in zebrafish also suggested that Axin1iEC−OE led to CNS vascular regression and impaired maturation but not inhibition of ongoing angiogenesis within the CNS. Transcriptomic profiling of isolated, β-catenin signaling-deficient endothelial cells during early blood-brain barrier–development (E11.5) revealed ECM (extracellular matrix) proteins as one of the most severely deregulated clusters. Among the 20 genes constituting the forebrain endothelial cell-specific response signature, 8 (Adamtsl2, Apod, Ctsw, Htra3, Pglyrp1, Spock2, Ttyh2, and Wfdc1) encoded bona fide ECM proteins. This specific β-catenin-responsive ECM signature was also repressed in Axin1iEC−OE and endothelial cell-specific β-catenin–knockout mice (Ctnnb1-KOiEC) during initial blood-brain barrier maturation (E14.5), consistent with an important role of Wnt/β-catenin signaling in orchestrating the development of the forebrain vascular ECM. Conclusions— These results suggest a novel mechanism of establishing a CNS endothelium-specific ECM signature downstream of Wnt-β-catenin that impact spatiotemporally on blood-brain barrier differentiation during forebrain vessel development.
Collapse
Affiliation(s)
- Lasse D Jensen
- From the Department of Medical and Health Sciences, Linköpings Universitet, Linköping, Sweden (L.D.J.)
| | - Belma Hot
- Department of Physiology and Pharmacology (B.H., V.M.L., J.K.), Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.)
| | - Daniel Ramsköld
- Department of Medicine, Solna (D.R.), Karolinska Institutet, Stockholm, Sweden.,Department of Cell and Molecular Biology (D.R., S.G., R.S., J.F.), Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.)
| | - Raoul F V Germano
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, Université libre de Bruxelles, Belgium (R.F.V.G., B.V.)
| | - Chika Yokota
- Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.).,Department of Biochemistry and Biophysics, Stockholm University, Sweden (C.Y.)
| | - Sarantis Giatrellis
- Department of Cell and Molecular Biology (D.R., S.G., R.S., J.F.), Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology (B.H., V.M.L., J.K.), Karolinska Institutet, Stockholm, Sweden
| | - Dirk Hubmacher
- Orthopaedic Research Laboratories, Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY (D.H.)
| | - Minerva X Li
- Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.).,Department of Clinical Sciences, Lunds Universitet, Sweden (M.X.L.)
| | - Mike Hupe
- Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.).,Developmental Biochemistry, Theodor Boveri Institute (Biocenter), University of Wuerzburg, Germany (M.H.)
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco (T.D.A.)
| | - Rickard Sandberg
- Department of Cell and Molecular Biology (D.R., S.G., R.S., J.F.), Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.)
| | - Jonas Frisén
- Department of Cell and Molecular Biology (D.R., S.G., R.S., J.F.), Karolinska Institutet, Stockholm, Sweden
| | - Marta Trusohamn
- Department of Medical Biochemistry and Biophysics (M.T., A.M., J.W.-K., D.N.), Karolinska Institutet, Stockholm, Sweden
| | - Agnieszka Martowicz
- Department of Medical Biochemistry and Biophysics (M.T., A.M., J.W.-K., D.N.), Karolinska Institutet, Stockholm, Sweden
| | - Joanna Wisniewska-Kruk
- Department of Medical Biochemistry and Biophysics (M.T., A.M., J.W.-K., D.N.), Karolinska Institutet, Stockholm, Sweden
| | - Daniel Nyqvist
- Department of Medical Biochemistry and Biophysics (M.T., A.M., J.W.-K., D.N.), Karolinska Institutet, Stockholm, Sweden
| | - Ralf H Adams
- Department of Tissue Morphogenesis Max-Planck-Institute for Molecular Biomedicine, University of Münster, Faculty of Medicine, Germany (R.H.A.)
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland Clinic Foundation (S.S.A.)
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, Université libre de Bruxelles, Belgium (R.F.V.G., B.V.).,Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Belgium (B.V.)
| | - Jan M Stenman
- Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.)
| | - Julianna Kele
- Department of Physiology and Pharmacology (B.H., V.M.L., J.K.), Karolinska Institutet, Stockholm, Sweden.,Ludwig Institute for Cancer Research Ltd, Stockholm, Sweden (B.H., D.R., C.Y., M.X.L., M.H., R.S., J.M.S., J.K.)
| |
Collapse
|
179
|
Chen-Roetling J, Cao Y, Peng D, Regan RF. Rapid loss of perihematomal cell viability in the collagenase intracerebral hemorrhage model. Brain Res 2019; 1711:91-96. [DOI: https:/doi.org/10.1016/j.brainres.2019.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
180
|
Kunze R, Marti HH. Angioneurins - Key regulators of blood-brain barrier integrity during hypoxic and ischemic brain injury. Prog Neurobiol 2019; 178:101611. [PMID: 30970273 DOI: 10.1016/j.pneurobio.2019.03.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
The loss of blood-brain barrier (BBB) integrity leading to vasogenic edema and brain swelling is a common feature of hypoxic/ischemic brain diseases such as stroke, but is also central to the etiology of other CNS disorders. In the past decades, numerous proteins, belonging to the family of angioneurins, have gained increasing attention as potential therapeutic targets for ischemic stroke, but also other CNS diseases attributed to BBB dysfunction. Angioneurins encompass mediators that affect both neuronal and vascular function. Recently, increasing evidence has been accumulated that certain angioneurins critically determine disease progression and outcome in stroke among others through multifaceted effects on the compromised BBB. Here, we will give a concise overview about the family of angioneurins. We further describe the most important cellular and molecular components that contribute to structural integrity and low permeability of the BBB under steady-state conditions. We then discuss BBB alterations in ischemic stroke, and highlight underlying cellular and molecular mechanisms. For the most prominent angioneurin family members including vascular endothelial growth factors, angiopoietins, platelet-derived growth factors and erythropoietin, we will summarize current scientific literature from experimental studies in animal models, and if available from clinical trials, on the following points: (i) spatiotemporal expression of these factors in the healthy and hypoxic/ischemic CNS, (ii) impact of loss- or gain-of-function during cerebral hypoxia/ischemia for BBB integrity and beyond, and (iii) potential underlying molecular mechanisms. Moreover, we will highlight novel therapeutic strategies based on the activation of endogenous angioneurins that might improve BBB dysfuntion during ischemic stroke.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany.
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Heidelberg University, Germany
| |
Collapse
|
181
|
Ysrayl BB, Balasubramaniam M, Albert I, Villalta F, Pandhare J, Dash C. A Novel Role of Prolidase in Cocaine-Mediated Breach in the Barrier of Brain Microvascular Endothelial Cells. Sci Rep 2019; 9:2567. [PMID: 30796241 PMCID: PMC6385491 DOI: 10.1038/s41598-018-37495-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Cocaine use is associated with breach in the blood brain barrier (BBB) and increased HIV-1 neuro-invasion. We show that the cellular enzyme "Prolidase" plays a key role in cocaine-induced disruption of the BBB. We established a barrier model to mimic the BBB by culturing human brain microvascular endothelial cells (HBMECs) in transwell inserts. In this model, cocaine treatment enhanced permeability of FITC-dextran suggesting a breach in the barrier. Interestingly, cocaine treatment increased the activity of matrix metallo-proteinases that initiate degradation of the BBB-associated collagen. Cocaine exposure also induced prolidase expression and activity in HBMECs. Prolidase catalyzes the final and rate-limiting step of collagen degradation during BBB remodeling. Knock-down of prolidase abrogated cocaine-mediated increased permeability suggesting a direct role of prolidase in BBB breach. To decipher the mechanism by which cocaine regulates prolidase, we probed the inducible nitric oxide synthase (iNOS) mediated phosphorylation of prolidase since mRNA levels of the protein were not altered upon cocaine treatment. We observed increased iNOS expression concurrent with increased prolidase phosphorylation in cocaine treated cells. Subsequently, inhibition of iNOS decreased prolidase phosphorylation and reduced cocaine-mediated permeability. Finally, cocaine treatment increased transmigration of monocytic cells through the HBMEC barrier. Knock-down of prolidase reduced cocaine-mediated monocyte transmigration, establishing a key role of prolidase in cocaine-induced breach in endothelial cell barrier.
Collapse
Affiliation(s)
- Binah Baht Ysrayl
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Ife Albert
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA
| | - Fernando Villalta
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA.
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA.
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA.
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA.
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, Tennessee, USA.
- Center for Molecular and Behavioral Neurosciences, Meharry Medical College, Nashville, Tennessee, USA.
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee, USA.
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, Tennessee, USA.
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, Tennessee, USA.
| |
Collapse
|
182
|
Nirwane A, Johnson J, Nguyen B, Miner JH, Yao Y. Mural cell-derived laminin-α5 plays a detrimental role in ischemic stroke. Acta Neuropathol Commun 2019; 7:23. [PMID: 30777135 PMCID: PMC6378751 DOI: 10.1186/s40478-019-0676-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 02/10/2019] [Indexed: 12/12/2022] Open
Abstract
At the blood-brain barrier (BBB), laminin-α5 is predominantly synthesized by endothelial cells and mural cells. Endothelial laminin-α5 is dispensable for BBB maintenance under homeostatic conditions but inhibits inflammatory cell extravasation in pathological conditions. Whether mural cell-derived laminin-α5 is involved in vascular integrity regulation, however, remains unknown. To answer this question, we generated transgenic mice with laminin-α5 deficiency in mural cells (α5-PKO). Under homeostatic conditions, no defects in BBB integrity and cerebral blood flow (CBF) were observed in α5-PKO mice, suggesting that mural cell-derived laminin-α5 is dispensable for BBB maintenance and CBF regulation under homeostatic conditions. After ischemia-reperfusion (MCAO) injury, however, α5-PKO mice displayed less severe neuronal injury, including reduced infarct volume, decreased neuronal death, and improved neurological function. In addition, α5-PKO mice also showed attenuated vascular damage (milder BBB disruption, reduced inflammatory cell infiltration, decreased brain edema, and diminished hemorrhagic transformation). Mechanistic studies revealed less severe tight junction protein (TJP) loss and pericyte coverage reduction in α5-PKO mice after ischemia-reperfusion injury, indicating that the attenuated ischemic injury in α5-PKO mice is possibly due to less severe vascular damage. These findings suggest that mural cell-derived laminin-α5 plays a detrimental role in ischemic stroke and that inhibiting its signaling may have a neuroprotective effect.
Collapse
|
183
|
Pétrault M, Casolla B, Ouk T, Cordonnier C, Bérézowski V. Cerebral microbleeds: Beyond the macroscope. Int J Stroke 2019; 14:468-475. [DOI: 10.1177/1747493019830594] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
While being increasingly recognized in clinical routine, brain microbleeds remain a puzzling finding for physicians. These small dot-like lesions are thought to be old perivascular collections of hemosiderin deposits. They can be found in different neurological settings such as cerebrovascular or neurodegenerative diseases. While their microscopic size would suggest considering these lesions as anecdotal, they are now regarded as biomarkers of severity of an underlying cerebrovascular disease. Their natural history and the interactions with surrounding brain cells remain unknown. However, their presence may impact therapeutic decisions. Deciphering the biological mechanisms leading to, or following microbleeds would enable us to address a key question: do microbleeds arise and impact the surrounding parenchyma like a miniature version of intracerebral hemorrhages or do they represent a different kind of injury? We hereby discuss, based on both clinical and experimental literature, the gap between the definition of microbleeds coming from neuroimaging and the pathophysiological hypotheses raised from histopathological and experimental data. Our analysis supports the need for a convergent effort from clinicians and basic scientists to go beyond the current “macro” view and disclose the cellular and molecular insights of these cerebral hemorrhagic microlesions.
Collapse
Affiliation(s)
- Maud Pétrault
- Department of Medical Pharmacology, Univ Lille, Inserm U1171-Degenerative and Vascular Cognitive Disorders, CHU Lille, Lille, France
| | - Barbara Casolla
- Department of Neurology, Univ Lille, Inserm U1171-Degenerative and Vascular Cognitive Disorders, CHU Lille, Lille, France
| | - Thavarak Ouk
- Department of Medical Pharmacology, Univ Lille, Inserm U1171-Degenerative and Vascular Cognitive Disorders, CHU Lille, Lille, France
| | - Charlotte Cordonnier
- Department of Neurology, Univ Lille, Inserm U1171-Degenerative and Vascular Cognitive Disorders, CHU Lille, Lille, France
| | - Vincent Bérézowski
- Department of Medical Pharmacology, Univ Lille, Inserm U1171-Degenerative and Vascular Cognitive Disorders, CHU Lille, Lille, France
- Univ Artois, Lens, France
| |
Collapse
|
184
|
Małkiewicz MA, Szarmach A, Sabisz A, Cubała WJ, Szurowska E, Winklewski PJ. Blood-brain barrier permeability and physical exercise. J Neuroinflammation 2019; 16:15. [PMID: 30678702 PMCID: PMC6345022 DOI: 10.1186/s12974-019-1403-x] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
In this narrative review, a theoretical framework on the crosstalk between physical exercise and blood-brain barrier (BBB) permeability is presented. We discuss the influence of physical activity on the factors affecting BBB permeability such as systemic inflammation, the brain renin-angiotensin and noradrenergic systems, central autonomic function and the kynurenine pathway. The positive role of exercise in multiple sclerosis and Alzheimer’s disease is described. Finally, the potential role of conditioning as well as the effect of exercise on BBB tight junctions is outlined. There is a body of evidence that regular physical exercise diminishes BBB permeability as it reinforces antioxidative capacity, reduces oxidative stress and has anti-inflammatory effects. It improves endothelial function and might increase the density of brain capillaries. Thus, physical training can be emphasised as a component of prevention programs developed for patients to minimise the risk of the onset of neuroinflammatory diseases as well as an augmentation of existing treatment. Unfortunately, despite a sound theoretical background, it remains unclear as to whether exercise training is effective in modulating BBB permeability in several specific diseases. Further research is needed as the impact of exercise is yet to be fully elucidated.
Collapse
Affiliation(s)
- Marta A Małkiewicz
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland.,Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Arkadiusz Szarmach
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Sabisz
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Wiesław J Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdansk, Gdansk, Poland
| | - Edyta Szurowska
- 2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland
| | - Paweł J Winklewski
- Department of Human Physiology, Faculty of Health Sciences, Medical University of Gdansk, Tuwima Str. 15, 80-210, Gdansk, Poland. .,2-nd Department of Radiology, Faculty of Health Sciences, Medical University of Gdansk, Gdansk, Poland. .,Department of Clinical Anatomy and Physiology, Faculty of Health Sciences, Pomeranian University of Slupsk, Slupsk, Poland.
| |
Collapse
|
185
|
Keeley DP, Sherwood DR. Tissue linkage through adjoining basement membranes: The long and the short term of it. Matrix Biol 2019; 75-76:58-71. [PMID: 29803937 PMCID: PMC6252152 DOI: 10.1016/j.matbio.2018.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 01/01/2023]
Abstract
Basement membranes (BMs) are thin dense sheets of extracellular matrix that surround most tissues. When the BMs of neighboring tissues come into contact, they usually slide along one another and act to separate tissues and organs into distinct compartments. However, in certain specialized regions, the BMs of neighboring tissues link, helping to bring tissues together. These BM connections can be transient, such as during tissue fusion events in development, or long-term, as with adult tissues involved with filtration, including the blood brain barrier and kidney glomerulus. The transitory nature of these connections in development and the complexity of tissue filtration systems in adults have hindered the understanding of how juxtaposed BMs fasten together. The recent identification of a BM-BM adhesion system in C. elegans, termed B-LINK (BM linkage), however, is revealing cellular and extracellular matrix components of a nascent tissue adhesion system. We discuss insights gained from studying the B-LINK tissue adhesion system in C. elegans, compare this adhesion with other BM-BM connections in Drosophila and vertebrates, and outline important future directions towards elucidating this fascinating and poorly understood mode of adhesion that joins neighboring tissues.
Collapse
Affiliation(s)
- Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, Box 90338, Durham, NC 27708, USA
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Box 90338, Durham, NC 27708, USA.
| |
Collapse
|
186
|
Abstract
Located at the interface of the circulation system and the CNS, the basement membrane (BM) is well positioned to regulate blood-brain barrier (BBB) integrity. Given the important roles of BBB in the development and progression of various neurological disorders, the BM has been hypothesized to contribute to the pathogenesis of these diseases. After stroke, a cerebrovascular disease caused by rupture (hemorrhagic) or occlusion (ischemic) of cerebral blood vessels, the BM undergoes constant remodeling to modulate disease progression. Although an association between BM dissolution and stroke is observed, how each individual BM component changes after stroke and how these components contribute to stroke pathogenesis are mostly unclear. In this review, I first briefly introduce the composition of the BM in the brain. Next, the functions of the BM and its major components in BBB maintenance under homeostatic conditions are summarized. Furthermore, the roles of the BM and its major components in the pathogenesis of hemorrhagic and ischemic stroke are discussed. Last, unsolved questions and potential future directions are described. This review aims to provide a comprehensive reference for future studies, stimulate the formation of new ideas, and promote the generation of new genetic tools in the field of BM/stroke research.
Collapse
Affiliation(s)
- Yao Yao
- Yao Yao, Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 340 Pharmacy South Building, 250 West Green Street, Athens, GA 30602, USA.
| |
Collapse
|
187
|
Microvascular networks in the area of the auditory peripheral nervous system. Hear Res 2019; 371:105-116. [DOI: 10.1016/j.heares.2018.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/14/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
|
188
|
Sweeney MD, Zhao Z, Montagne A, Nelson AR, Zlokovic BV. Blood-Brain Barrier: From Physiology to Disease and Back. Physiol Rev 2019; 99:21-78. [PMID: 30280653 PMCID: PMC6335099 DOI: 10.1152/physrev.00050.2017] [Citation(s) in RCA: 1315] [Impact Index Per Article: 219.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 04/17/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) prevents neurotoxic plasma components, blood cells, and pathogens from entering the brain. At the same time, the BBB regulates transport of molecules into and out of the central nervous system (CNS), which maintains tightly controlled chemical composition of the neuronal milieu that is required for proper neuronal functioning. In this review, we first examine molecular and cellular mechanisms underlying the establishment of the BBB. Then, we focus on BBB transport physiology, endothelial and pericyte transporters, and perivascular and paravascular transport. Next, we discuss rare human monogenic neurological disorders with the primary genetic defect in BBB-associated cells demonstrating the link between BBB breakdown and neurodegeneration. Then, we review the effects of genes underlying inheritance and/or increased susceptibility for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and amyotrophic lateral sclerosis (ALS) on BBB in relation to other pathologies and neurological deficits. We next examine how BBB dysfunction relates to neurological deficits and other pathologies in the majority of sporadic AD, PD, and ALS cases, multiple sclerosis, other neurodegenerative disorders, and acute CNS disorders such as stroke, traumatic brain injury, spinal cord injury, and epilepsy. Lastly, we discuss BBB-based therapeutic opportunities. We conclude with lessons learned and future directions, with emphasis on technological advances to investigate the BBB functions in the living human brain, and at the molecular and cellular level, and address key unanswered questions.
Collapse
Affiliation(s)
- Melanie D Sweeney
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Amy R Nelson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California , Los Angeles, California ; and Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California , Los Angeles, California
| |
Collapse
|
189
|
Chen Y, Huang A, Ao W, Wang Z, Yuan J, Song Q, Wei D, Ye H. Proteomic analysis of serum proteins from HIV/AIDS patients with Talaromyces marneffei infection by TMT labeling-based quantitative proteomics. Clin Proteomics 2018; 15:40. [PMID: 30598657 PMCID: PMC6302400 DOI: 10.1186/s12014-018-9219-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Talaromyces marneffei (TM) is an emerging pathogenic fungus that can cause a fatal systemic mycosis in patients infected with human immunodeficiency virus (HIV). Although global awareness regarding HIV/TM coinfection is increasing little is known about the mechanism that mediates the rapid progression to HIV/AIDS disease in coinfected individuals. The aim of this study was to analyze the serum proteome of HIV/TM coinfected patients and to identify the associated protein biomarkers for TM in patients with HIV/AIDS. METHODS We systematically used multiplexed isobaric tandem mass tag labeling combined with liquid chromatography mass spectrometry (LC-MS/MS) to screen for differentially expressed proteins in the serum samples from HIV/TM-coinfected patients. RESULTS Of a total data set that included 1099 identified proteins, approximately 86% of the identified proteins were quantified. Among them, 123 proteins were at least 1.5-fold up-or downregulated in the serum between HIV/TM-coinfected and HIV-mono-infected patients. Furthermore, our results indicate that two selected proteins (IL1RL1 and THBS1) are potential biomarkers for distinguishing HIV/TM-coinfected patients. CONCLUSIONS This is the first report to provide a global proteomic profile of serum samples from HIV/TM-coinfected patients. Our data provide insights into the proteins that are involved as host response factors during infection. These data shed new light on the molecular mechanisms that are dysregulated and contribute to the pathogenesis of HIV/TM coinfection. IL1RL1 and THBS1 are promising diagnostic markers for HIV/TM-coinfected patients although further large-scale studies are needed. Thus, quantitative proteomic analysis revealed molecular differences between the HIV/TM-coinfected and HIV-mono-infected individuals, and might provide fundamental information for further detailed investigations.
Collapse
Affiliation(s)
- Yahong Chen
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
| | - Aiqiong Huang
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
| | - Wen Ao
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
| | - Zhengwu Wang
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
| | - Jinjin Yuan
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
| | - Qing Song
- Shanxi Institute of Flexible Electronics, Northwestern Polytechnical University, 127 West Youyi Road, Xi’an, 710072 People’s Republic of China
| | - Dahai Wei
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- The First Affiliated Hospital of Jiaxing University, 1882 Zhonghuan Road, Jiaxing, 314001 People’s Republic of China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025 People’s Republic of China
| | - Hanhui Ye
- Mengchao Hepatobiliary Hospital of Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
- Fuzhou Infectious Disease Hospital, Fujian Medical University, 312 Xihong Road, Fuzhou, 350025 Fujian Province People’s Republic of China
| |
Collapse
|
190
|
Xu L, Nirwane A, Yao Y. Basement membrane and blood-brain barrier. Stroke Vasc Neurol 2018; 4:78-82. [PMID: 31338215 PMCID: PMC6613871 DOI: 10.1136/svn-2018-000198] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 11/16/2018] [Indexed: 12/24/2022] Open
Abstract
The blood–brain barrier (BBB) is a highly complex and dynamic structure, mainly composed of brain microvascular endothelial cells, pericytes, astrocytes and the basement membrane (BM). The vast majority of BBB research focuses on its cellular constituents. Its non-cellular component, the BM, on the other hand, is largely understudied due to its intrinsic complexity and the lack of research tools. In this review, we focus on the role of the BM in BBB integrity. We first briefly introduce the biochemical composition and structure of the BM. Next, the biological functions of major components of the BM in BBB formation and maintenance are discussed. Our goal is to provide a concise overview on how the BM contributes to BBB integrity.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, USA
| | - Abhijit Nirwane
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
191
|
Edwards DN, Bix GJ. Roles of blood-brain barrier integrins and extracellular matrix in stroke. Am J Physiol Cell Physiol 2018; 316:C252-C263. [PMID: 30462535 DOI: 10.1152/ajpcell.00151.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemicstroke is a leading cause of death and disability in the United States, but recent advances in treatments [i.e., endovascular thrombectomy and tissue plasminogen activator (t-PA)] that target the stroke-causing blood clot, while improving overall stroke mortality rates, have had much less of an impact on overall stroke morbidity. This may in part be attributed to the lack of therapeutics targeting reperfusion-induced injury after the blood clot has been removed, which, if left unchecked, can expand injury from its core into the surrounding at risk tissue (penumbra). This occurs in two phases of increased permeability of the blood-brain barrier, a physical barrier that under physiologic conditions regulates brain influx and efflux of substances and consists of tight junction forming endothelial cells (and transporter proteins), astrocytes, pericytes, extracellular matrix, and their integrin cellular receptors. During, embryonic development, maturity, and following stroke reperfusion, cerebral vasculature undergoes significant changes including changes in expression of integrins and degradation of surrounding extracellular matrix. Integrins, heterodimers with α and β subunits, and their extracellular matrix ligands, a collection of proteoglycans, glycoproteins, and collagens, have been modestly studied in the context of stroke compared with other diseases (e.g., cancer). In this review, we describe the effect that various integrins and extracellular matrix components have in embryonic brain development, and how this changes in both maturity and in the poststroke environment. Particular focus will be on how these changes in integrins and the extracellular matrix affect blood-brain barrier components and their potential as diagnostic and therapeutic targets for ischemic stroke.
Collapse
Affiliation(s)
- Danielle N Edwards
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky.,Department of Neuroscience, University of Kentucky , Lexington, Kentucky
| | - Gregory J Bix
- Sanders-Brown Center on Aging, University of Kentucky , Lexington, Kentucky.,Department of Neuroscience, University of Kentucky , Lexington, Kentucky.,Department of Neurology, University of Kentucky , Lexington, Kentucky.,Department of Neurosurgery, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
192
|
Clément T, Rodriguez-Grande B, Badaut J. Aquaporins in brain edema. J Neurosci Res 2018; 98:9-18. [DOI: 10.1002/jnr.24354] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 10/15/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Tifenn Clément
- CNRS UMR 5287, INCIA, University of Bordeaux; Bordeaux France
| | | | - Jérôme Badaut
- CNRS UMR 5287, INCIA, University of Bordeaux; Bordeaux France
- Department of Basic Science; Loma Linda University School of Medicine; Loma Linda California
| |
Collapse
|
193
|
Segarra M, Aburto MR, Cop F, Llaó-Cid C, Härtl R, Damm M, Bethani I, Parrilla M, Husainie D, Schänzer A, Schlierbach H, Acker T, Mohr L, Torres-Masjoan L, Ritter M, Acker-Palmer A. Endothelial Dab1 signaling orchestrates neuro-glia-vessel communication in the central nervous system. Science 2018; 361:361/6404/eaao2861. [PMID: 30139844 DOI: 10.1126/science.aao2861] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 07/05/2018] [Indexed: 12/17/2022]
Abstract
The architecture of the neurovascular unit (NVU) is controlled by the communication of neurons, glia, and vascular cells. We found that the neuronal guidance cue reelin possesses proangiogenic activities that ensure the communication of endothelial cells (ECs) with the glia to control neuronal migration and the establishment of the blood-brain barrier in the mouse brain. Apolipoprotein E receptor 2 (ApoER2) and Disabled1 (Dab1) expressed in ECs are required for vascularization of the retina and the cerebral cortex. Deletion of Dab1 in ECs leads to a reduced secretion of laminin-α4 and decreased activation of integrin-β1 in glial cells, which in turn control neuronal migration and barrier properties of the NVU. Thus, reelin signaling in the endothelium is an instructive and integrative cue essential for neuro-glia-vascular communication.
Collapse
Affiliation(s)
- Marta Segarra
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Maria R Aburto
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany.,Focus Program Translational Neurosciences, University of Mainz, D-55131 Mainz, Germany
| | - Florian Cop
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Cecília Llaó-Cid
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Ricarda Härtl
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Miriam Damm
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany.,Focus Program Translational Neurosciences, University of Mainz, D-55131 Mainz, Germany
| | - Ioanna Bethani
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Marta Parrilla
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany.,Max Planck Institute for Brain Research, D-60438 Frankfurt am Main, Germany
| | - Dewi Husainie
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany.,Max Planck Institute for Brain Research, D-60438 Frankfurt am Main, Germany
| | - Anne Schänzer
- Institute of Neuropathology, University of Giessen, D-35392 Giessen, Germany
| | - Hannah Schlierbach
- Institute of Neuropathology, University of Giessen, D-35392 Giessen, Germany
| | - Till Acker
- Institute of Neuropathology, University of Giessen, D-35392 Giessen, Germany
| | - Laura Mohr
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Laia Torres-Masjoan
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Mathias Ritter
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany. .,Focus Program Translational Neurosciences, University of Mainz, D-55131 Mainz, Germany.,Max Planck Institute for Brain Research, D-60438 Frankfurt am Main, Germany
| |
Collapse
|
194
|
Da Mesquita S, Fu Z, Kipnis J. The Meningeal Lymphatic System: A New Player in Neurophysiology. Neuron 2018; 100:375-388. [PMID: 30359603 PMCID: PMC6268162 DOI: 10.1016/j.neuron.2018.09.022] [Citation(s) in RCA: 288] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/30/2018] [Accepted: 09/11/2018] [Indexed: 12/24/2022]
Abstract
The nature of fluid dynamics within the brain parenchyma is a focus of intensive research. Of particular relevance is its participation in diseases associated with protein accumulation and aggregation in the brain, such as Alzheimer's disease (AD). The meningeal lymphatic vessels have recently been recognized as an important player in the complex circulation and exchange of soluble contents between the cerebrospinal fluid (CSF) and the interstitial fluid (ISF). In aging mammals, for example, impaired functioning of the meningeal lymphatic vessels can lead to accelerated accumulation of toxic amyloid beta protein in the brain parenchyma, thus aggravating AD-related pathology. Given that meningeal lymphatic vessels are functionally linked to paravascular influx/efflux of the CSF/ISF, and in light of recent findings that certain cytokines, classically perceived as immune molecules, exert neuromodulatory effects, it is reasonable to suggest that the activity of meningeal lymphatics could alter the accessibility of CSF-borne immune neuromodulators to the brain parenchyma, thereby altering their effects on the brain. Accordingly, in this Perspective we propose that the meningeal lymphatic system can be viewed as a novel player in neurophysiology.
Collapse
Affiliation(s)
- Sandro Da Mesquita
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| | - Zhongxiao Fu
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
195
|
Labus J, Wöltje K, Stolte KN, Häckel S, Kim KS, Hildmann A, Danker K. IL-1β promotes transendothelial migration of PBMCs by upregulation of the FN/α 5β 1 signalling pathway in immortalised human brain microvascular endothelial cells. Exp Cell Res 2018; 373:99-111. [PMID: 30342992 DOI: 10.1016/j.yexcr.2018.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 09/29/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is often associated with pathological changes in the function of the blood-brain barrier (BBB) caused by disassembly of tight and adherens junctions that under physiological conditions are important for the maintenance of the BBB integrity. Consequently, in inflammation the BBB becomes dysfunctional, facilitating leukocyte traversal of the barrier and accumulation of immune cells within the brain. The extracellular matrix (ECM) also contributes to BBB integrity but the significance of the main ECM receptors, the β1 integrins also expressed on endothelial cells, is less well understood. To evaluate whether β1 integrin function is affected during inflammation and impacts barrier function, we used a transformed human brain microvascular endothelial cell (THBMEC)-based Interleukin 1β (IL-1β)-induced inflammatory in vitro BBB model. We demonstrate that IL-1β increases cell-matrix adhesion and induces a redistribution of active β1 integrins to the basal surface. In particular, binding of α5β1 integrin to its ligand fibronectin is enhanced and α5β1 integrin-dependent signalling is upregulated. Additionally, localisation of the tight junction protein claudin-5 is altered. Blockade of the α5β1 integrin reduces the IL-1β-induced transendothelial migration of peripheral blood mononuclear cells (PBMCs). These data imply that IL-1β-induced inflammation not only destabilizes tight junctions but also increases α5β1 integrin-dependent cell-matrix adhesion to fibronectin.
Collapse
Affiliation(s)
- Josephine Labus
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117 Berlin, Germany
| | - Kerstin Wöltje
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117 Berlin, Germany
| | - Kim Natalie Stolte
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117 Berlin, Germany
| | - Sonja Häckel
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117 Berlin, Germany
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 200 North Wolfe Street, 21287 Baltimore, USA
| | - Annette Hildmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117 Berlin, Germany
| | - Kerstin Danker
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
196
|
Davis Armstrong NM, Chen WM, Brewer MS, Williams SR, Sale MM, Worrall BB, Keene KL. Epigenome-Wide Analyses Identify Two Novel Associations With Recurrent Stroke in the Vitamin Intervention for Stroke Prevention Clinical Trial. Front Genet 2018; 9:358. [PMID: 30237808 PMCID: PMC6135883 DOI: 10.3389/fgene.2018.00358] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022] Open
Abstract
DNA methylation, a well-characterized epigenetic modification that is influenced by both environment and genetic variation, has previously been implicated in a number of complex diseases, including cardiovascular disease and stroke. The goal of this study was to evaluate epigenome-wide associations with recurrent stroke and the folate one-carbon metabolism-related trait, plasma homocysteine (hcy). Differential methylation analyses were performed on 473,864 autosomal CpG loci, using Illumina HumanMethylation 450K array data in 180 ischemic stroke cases from the Vitamin Intervention for Stroke Prevention (VISP) clinical trial. Linear regression was used to assess associations between number of strokes prior to VISP enrollment and measures of hcy with degree of methylation (β-values), while logistic regression was used to evaluate recurrent stroke status and incident recurrent stroke associations. All regression analyses were stratified by race. Two differentially methylated CpG sites exceeded epigenome-wide significance (p ≤ 1.055 × 10−7) for prior number of strokes (PNS) in European Americans. The top locus, cg22812874, was located in the ankyrin repeat and SOCS box containing 10 gene (ASB10; p = 3.4 × 10−9; β = −0.0308; 95% CI = −0.040, −0.002). Methylation locus cg00340919, located in an intron of the tetratricopeptide repeat domain 37 gene, was also statistically significant (TTC37; p = 8.74 × 10−8; β = −0.0517; 95% CI = −0.069, −0.034). An additional 138 CpG sites met our threshold for suggestive significance (p ≤ 5 × 10−5). We evaluated DNA methylation associated with recurrent stroke and hcy phenotypes across the epigenome. Hypermethylation at two CpG sites located in ASB10 and TTC37 was associated with fewer strokes prior to VISP enrollment. Our findings present a foundation for additional epigenome-wide studies, as well as mechanistic studies into epigenetic marks that influence recurrent stroke risk.
Collapse
Affiliation(s)
| | - Wei-Min Chen
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States.,Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | - Michael S Brewer
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Stephen R Williams
- Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Michèle M Sale
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States.,Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | - Bradford B Worrall
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States.,Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Keith L Keene
- Department of Biology, East Carolina University, Greenville, NC, United States.,Center for Health Disparities, East Carolina University, Greenville, NC, United States
| |
Collapse
|
197
|
Xiang J, Andjelkovic AV, Zhou N, Hua Y, Xi G, Wang MM, Keep RF. Is there a central role for the cerebral endothelium and the vasculature in the brain response to conditioning stimuli? CONDITIONING MEDICINE 2018; 1:220-232. [PMID: 30906928 PMCID: PMC6426135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
A variety of conditioning stimuli (e.g. ischemia or hypoxia) can protect against stroke-induced brain injury. While most attention has focused on the effects of conditioning on parenchymal injury, there is considerable evidence that such stimuli also protect the cerebrovasculature, including the blood-brain barrier. This review summarizes the data on the cerebrovascular effects of ischemic/hypoxic pre-, per- and post-conditioning and the mechanisms involved in protection. It also addresses some important questions: Are the cerebrovascular effects of conditioning just secondary to reduced parenchymal injury? How central is endothelial conditioning to overall brain protection? For example, is endothelial conditioning sufficient or necessary for the induction of brain protection against stroke? Is the endothelium crucial as a sensor/transducer of conditioning stimuli?
Collapse
Affiliation(s)
- Jianming Xiang
- Department of Neurosurgery, Medical School, University of Michigan
| | - Anuska V. Andjelkovic
- Department of Neurosurgery, Medical School, University of Michigan
- Department of Pathology, Medical School, University of Michigan
| | - Ningna Zhou
- Department of Neurosurgery, Medical School, University of Michigan
- Department of Pharmacology, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Ya Hua
- Department of Neurosurgery, Medical School, University of Michigan
| | - Guohua Xi
- Department of Neurosurgery, Medical School, University of Michigan
| | - Michael M. Wang
- Department of Neurology, Medical School, University of Michigan
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Richard F. Keep
- Department of Neurosurgery, Medical School, University of Michigan
| |
Collapse
|
198
|
Chen X, Patra A, Sadowska GB, Stonestreet BS. Ischemic-Reperfusion Injury Increases Matrix Metalloproteinases and Tissue Metalloproteinase Inhibitors in Fetal Sheep Brain. Dev Neurosci 2018; 40:234-245. [PMID: 30048980 DOI: 10.1159/000489700] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/29/2018] [Indexed: 12/31/2022] Open
Abstract
Hypoxic-ischemic brain injury is a leading cause of neurodevelopmental morbidities in preterm and full-term infants. Blood-brain barrier dysfunction represents an important component of perinatal hypoxic-ischemic brain injury. The extracellular matrix (ECM) is a vital component of the blood-brain barrier. Matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) are important ECM components. They contribute to brain development, blood-brain barrier maintenance, and to regenerative and repair processes after hypoxic-ischemic brain injury. We hypothesized that ischemia at different durations of reperfusion affects the ECM protein composition of MMPs and TIMPs in the cerebral cortex of fetal sheep. Cerebral cortical samples were snap-frozen from sham control fetuses at 127 days of gestation and from fetuses after exposure to 30-min carotid occlusion and 4-, 24-, and 48-h of reperfusion. Protein expression of MMP-2, -8, -9, and -13 and TIMP-1, -2, -3, and -4 was measured by Western immunoblotting along with the gelatinolytic activity of MMP-2 and MMP-9 by zymography. The expression of MMP-8 was increased (Kruskal-Wallis, p = 0.04) in fetuses 48 h after ischemia. In contrast, changes were not observed in the protein expression of MMP-2, -9, or -13. The gelatinolytic activity of pro-MMP-2 was increased (ANOVA, p = 0.02, Tukey HSD, p = 0.05) 24 h after ischemia. TIMP-1 and -3 expression levels were also higher (TIMP-1, ANOVA, p = 0.003, Tukey HSD, p = 0.01; TIMP-3, ANOVA, p = 0.006, Tukey HSD, p = 0.01) 24 h after ischemia compared with both the sham controls and with fetuses exposed to 4 h of reperfusion. The changes in the expression of TIMP-1, -2, and -3 correlated with the changes in the MMP-8 and -13 protein expression. We speculate that regulation of MMP-8, MMP-13, and TIMPs contributes to ECM remodeling after is chemic-reperfusion injury in the fetal brain.
Collapse
|
199
|
Natarajan R, Mitchell CM, Harless N, Yamamoto BK. Cerebrovascular Injury After Serial Exposure to Chronic Stress and Abstinence from Methamphetamine Self-Administration. Sci Rep 2018; 8:10558. [PMID: 30002494 PMCID: PMC6043597 DOI: 10.1038/s41598-018-28970-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 07/04/2018] [Indexed: 01/19/2023] Open
Abstract
Cerebrovascular damage caused by either exposure to stress or the widely abused drug, methamphetamine (Meth) is known but stress and drug abuse frequently occur in tandem that may impact their individual cerebrovascular effects. This study examined their co-morbid cerebrovascular effects during abstinence from self-administered Meth after the exposure to chronic unpredictable stress (CUS). Exposure to CUS prior to unrestricted Meth self-administration had no effect on Meth intake in rats; however, the pro-inflammatory mediator cyclooxygenase-2 (COX-2) and the breakdown of cell-matrix adhesion protein β-dystroglycan in isolated cerebral cortical capillaries were increased after 3 days of abstinence and persisted for 7 days. These changes preceded decreases in occludin, a key structural protein component of the blood-brain barrier. The decrease in occludin was blocked by the COX-2 specific inhibitor nimesulide treatment during abstinence from Meth. The changes in COX-2, β-dystroglycan, and occludin were only evident following the serial exposure to stress and Meth but not after either one alone. These results suggest that stress and voluntary Meth intake can synergize and disrupt cerebrovasculature in a time-dependent manner during abstinence from chronic stress and Meth. Furthermore, COX-2 inhibition may be a viable pharmacological intervention to block vascular changes after Meth exposure.
Collapse
Affiliation(s)
- Reka Natarajan
- Department of Pharmacology and Toxicology, Indiana University School of Medicine 635 Barnhill Drive MS A401, Indianapolis, IN, 46202, USA
| | - Carmen M Mitchell
- Department of Pharmacology and Toxicology, Indiana University School of Medicine 635 Barnhill Drive MS A401, Indianapolis, IN, 46202, USA
| | - Nicole Harless
- Department of Neurosciences, University of Toledo College of Medicine 3000 Arlington Avenue MS 1007, Toledo, OH, 43614, Spain
| | - Bryan K Yamamoto
- Department of Pharmacology and Toxicology, Indiana University School of Medicine 635 Barnhill Drive MS A401, Indianapolis, IN, 46202, USA.
| |
Collapse
|
200
|
Thurgur H, Pinteaux E. Microglia in the Neurovascular Unit: Blood-Brain Barrier-microglia Interactions After Central Nervous System Disorders. Neuroscience 2018; 405:55-67. [PMID: 31007172 DOI: 10.1016/j.neuroscience.2018.06.046] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022]
Abstract
Over the past few decades, microglial cells have been regarded as the main executor of inflammation after acute and chronic central nervous system (CNS) disorders, responding rapidly to exogenous stimuli during acute trauma or infections, or signals released by cells undergoing cell death during conditions such as stroke, Alzheimer's disease (AD) and Parkinson's disease (PD). Barriers of the nervous system, and in particular the blood-brain barrier (BBB), play a key role in the normal physiological and cognitive functions of the brain. Being at the interface between the central and peripheral compartment, the BBB is regarded as a sensor of homeostasis, and any disruption within the brain or the systemic compartment triggers BBB dysfunction and neuroinflammation, both contributing to the pathogenesis of cerebrovascular disease. This involves a dynamic response mediated by all components of the neurovascular unit (NVU), and ongoing research suggests that BBB-microglia interaction is critical to dictate the microglial response to NVU injury. The present review aims to give an up-to-date account of the emerging critical role of BBB-microglia interactions during neuroinflammation, and how these could be targeted for the therapeutic treatment of major central inflammatory disease.
Collapse
Affiliation(s)
- Hannah Thurgur
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, United Kingdom
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, AV Hill Building, The University of Manchester, United Kingdom.
| |
Collapse
|