1
|
Hage Z, Madeira MM, Koliatsis D, Tsirka SE. Convergence of endothelial dysfunction, inflammation and glucocorticoid resistance in depression-related cardiovascular diseases. BMC Immunol 2024; 25:61. [PMID: 39333855 PMCID: PMC11428380 DOI: 10.1186/s12865-024-00653-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Major Depressive Disorder, or depression, has been extensively linked to dysregulated HPA axis function, chronic inflammation and cardiovascular diseases. While the former two have been studied in depth, the mechanistic connection between depression and cardiovascular disease is unclear. As major mediators of vascular homeostasis, vascular pathology and immune activity, endothelial cells represent an important player connecting the diseases. Exaggerated inflammation and glucocorticoid function are important topics to explore in the endothelial response to MDD. Glucocorticoid resistance in several cell types strongly promotes inflammatory signaling and results in worsened severity in many diseases. However, endothelial health and inflammation in chronic stress and depression are rarely considered from the perspective of glucocorticoid signaling and resistance. In this review, we aim to discuss (1) endothelial dysfunction in depression, (2) inflammation in depression, (3) general glucocorticoid resistance in depression and (4) endothelial glucocorticoid resistance in depression co-morbid inflammatory diseases. We will first describe vascular pathology, inflammation and glucocorticoid resistance separately in depression and then describe their potential interactions with one another in depression-relevant diseases. Lastly, we will hypothesize potential mechanisms by which glucocorticoid resistance in endothelial cells may contribute to vascular disease states in depressed people. Overall, endothelial-glucocorticoid signaling may play an important role in connecting depression and vascular pathology and warrants further study.
Collapse
Affiliation(s)
- Zachary Hage
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Miguel M Madeira
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Dimitris Koliatsis
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Stella E Tsirka
- Program in Molecular and Cellular Pharmacology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Department of Pharmacological Sciences, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
- Scholars in Biomedical Sciences Program, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
2
|
Jiang Y, Chen P, Zhao Y, Zhang Y. Association of Cadherin-Related Family Member 1 with Traumatic Brain Injury. Cell Mol Neurobiol 2024; 44:41. [PMID: 38656449 PMCID: PMC11043179 DOI: 10.1007/s10571-024-01476-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
The cadherin family plays a pivotal role in orchestrating synapse formation in the central nervous system. Cadherin-related family member 1 (CDHR1) is a photoreceptor-specific calmodulin belonging to the expansive cadherin superfamily. However, its role in traumatic brain injury (TBI) remains largely unknown. CDHR1 expression across various brain tissue sites was analyzed using the GSE104687 dataset. Employing a summary-data-based Mendelian Randomization (SMR) approach, integrated analyses were performed by amalgamating genome-wide association study abstracts from TBI with public data on expressed quantitative trait loci and DNA methylation QTL from both blood and diverse brain tissues. CDHR1 expression and localization in different brain tissues were meticulously delineated using western blotting, immunohistochemistry, and enzyme-linked immunosorbent assay. CDHR1 expression was consistently elevated in the TBI group compared to that in the sham group across multiple tissues. The inflammatory response emerged as a crucial biological mechanism, and pro-inflammatory and anti-inflammatory factors were not expressed in either group. Integrated SMR analyses encompassing both blood and brain tissues substantiated the heightened CDHR1 expression profiles, with methylation modifications emerging as potential contributing factors for increased TBI risk. This was corroborated by western blotting and immunohistochemistry, confirming augmented CDHR1 expression following TBI. This multi-omics-based genetic association study highlights the elevated TBI risk associated with CDHR1 expression coupled with putative methylation modifications. These findings provide compelling evidence for future targeted investigations and offer promising avenues for developing interventional therapies for TBI.
Collapse
Affiliation(s)
- Yong'An Jiang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
- Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Peng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
- Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - YangYang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
- Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yan Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
3
|
Parvez MM, Sadighi A, Ahn Y, Keller SF, Enoru JO. Uptake Transporters at the Blood-Brain Barrier and Their Role in Brain Drug Disposition. Pharmaceutics 2023; 15:2473. [PMID: 37896233 PMCID: PMC10610385 DOI: 10.3390/pharmaceutics15102473] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Uptake drug transporters play a significant role in the pharmacokinetic of drugs within the brain, facilitating their entry into the central nervous system (CNS). Understanding brain drug disposition is always challenging, especially with respect to preclinical to clinical translation. These transporters are members of the solute carrier (SLC) superfamily, which includes organic anion transporter polypeptides (OATPs), organic anion transporters (OATs), organic cation transporters (OCTs), and amino acid transporters. In this systematic review, we provide an overview of the current knowledge of uptake drug transporters in the brain and their contribution to drug disposition. Here, we also assemble currently available proteomics-based expression levels of uptake transporters in the human brain and their application in translational drug development. Proteomics data suggest that in association with efflux transporters, uptake drug transporters present at the BBB play a significant role in brain drug disposition. It is noteworthy that a significant level of species differences in uptake drug transporters activity exists, and this may contribute toward a disconnect in inter-species scaling. Taken together, uptake drug transporters at the BBB could play a significant role in pharmacokinetics (PK) and pharmacodynamics (PD). Continuous research is crucial for advancing our understanding of active uptake across the BBB.
Collapse
Affiliation(s)
- Md Masud Parvez
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Armin Sadighi
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Yeseul Ahn
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S Coulter St., Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Steve F. Keller
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| | - Julius O. Enoru
- Department of Quantitative, Translational & ADME Sciences (QTAS), AbbVie Biotherapeutics, San Francisco, CA 94080, USA; (M.M.P.)
| |
Collapse
|
4
|
Reinhold AK, Hartmannsberger B, Burek M, Rittner HL. Stabilizing the neural barrier - A novel approach in pain therapy. Pharmacol Ther 2023; 249:108484. [PMID: 37390969 DOI: 10.1016/j.pharmthera.2023.108484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/08/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Chronic and neuropathic pain are a widespread burden. Incomplete understanding of underlying pathomechanisms is one crucial factor for insufficient treatment. Recently, impairment of the blood nerve barrier (BNB) has emerged as one key aspect of pain initiation and maintenance. In this narrative review, we discuss several mechanisms and putative targets for novel treatment strategies. Cells such as pericytes, local mediators like netrin-1 and specialized proresolving mediators (SPMs), will be covered as well as circulating factors including the hormones cortisol and oestrogen and microRNAs. They are crucial in either the BNB or similar barriers and associated with pain. While clinical studies are still scarce, these findings might provide valuable insight into mechanisms and nurture development of therapeutic approaches.
Collapse
Affiliation(s)
- Ann-Kristin Reinhold
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Beate Hartmannsberger
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Malgorzata Burek
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany.
| |
Collapse
|
5
|
Hemorrhagic Cerebral Insults and Secondary Takotsubo Syndrome: Findings in a Novel In Vitro Model Using Human Blood Samples. Int J Mol Sci 2022; 23:ijms231911557. [PMID: 36232860 PMCID: PMC9569517 DOI: 10.3390/ijms231911557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Intracranial hemorrhage results in devastating forms of cerebral damage. Frequently, these results also present with cardiac dysfunction ranging from ECG changes to Takotsubo syndrome (TTS). This suggests that intracranial bleeding due to subarachnoid hemorrhage (SAH) disrupts the neuro-cardiac axis leading to neurogenic stress cardiomyopathy (NSC) of different degrees. Following this notion, SAH and secondary TTS could be directly linked, thus contributing to poor outcomes. We set out to test if blood circulation is the driver of the brain-heart axis by investigating serum samples of TTS patients. We present a novel in vitro model combining SAH and secondary TTS to mimic the effects of blood or serum, respectively, on blood-brain barrier (BBB) integrity using in vitro monolayers of an established murine model. We consistently demonstrated decreased monolayer integrity and confirmed reduced Claudin-5 and Occludin levels by RT-qPCR and Western blot and morphological reorganization of actin filaments in endothelial cells. Both tight junction proteins show a time-dependent reduction. Our findings highlight a faster and more prominent disintegration of BBB in the presence of TTS and support the importance of the bloodstream as a causal link between intracerebral bleeding and cardiac dysfunction. This may represent potential targets for future therapeutic inventions in SAH and TTS.
Collapse
|
6
|
Mapunda JA, Tibar H, Regragui W, Engelhardt B. How Does the Immune System Enter the Brain? Front Immunol 2022; 13:805657. [PMID: 35273596 PMCID: PMC8902072 DOI: 10.3389/fimmu.2022.805657] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple Sclerosis (MS) is considered the most frequent inflammatory demyelinating disease of the central nervous system (CNS). It occurs with a variable prevalence across the world. A rich armamentarium of disease modifying therapies selectively targeting specific actions of the immune system is available for the treatment of MS. Understanding how and where immune cells are primed, how they access the CNS in MS and how immunomodulatory treatments affect neuroinflammation requires a proper knowledge on the mechanisms regulating immune cell trafficking and the special anatomy of the CNS. The brain barriers divide the CNS into different compartments that differ with respect to their accessibility to cells of the innate and adaptive immune system. In steady state, the blood-brain barrier (BBB) limits immune cell trafficking to activated T cells, which can reach the cerebrospinal fluid (CSF) filled compartments to ensure CNS immune surveillance. In MS immune cells breach a second barrier, the glia limitans to reach the CNS parenchyma. Here we will summarize the role of the endothelial, epithelial and glial brain barriers in regulating immune cell entry into the CNS and which immunomodulatory treatments for MS target the brain barriers. Finally, we will explore current knowledge on genetic and environmental factors that may influence immune cell entry into the CNS during neuroinflammation in Africa.
Collapse
Affiliation(s)
| | - Houyam Tibar
- Medical School of Rabat, Mohamed 5 University, Rabat, Morocco.,Hôpital des spécialités de Rabat, Ibn Sina University Hospital of Rabat, Rabat, Morocco
| | - Wafa Regragui
- Medical School of Rabat, Mohamed 5 University, Rabat, Morocco.,Hôpital des spécialités de Rabat, Ibn Sina University Hospital of Rabat, Rabat, Morocco
| | | |
Collapse
|
7
|
Gu X, Ge L, Ren B, Fang Y, Li Y, Wang Y, Xu H. Glucocorticoids Promote Extracellular Matrix Component Remodeling by Activating YAP in Human Retinal Capillary Endothelial Cells. Front Cell Dev Biol 2022; 9:738341. [PMID: 34970541 PMCID: PMC8712730 DOI: 10.3389/fcell.2021.738341] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/14/2021] [Indexed: 11/13/2022] Open
Abstract
Remodeling of extracellular matrix (ECM) components of endothelial cells is the main cause of retinal vascular basement membrane (BM) thickening, which leads to the initiation and perpetuation of microvasculopathy of diabetic retinopathy (DR). Excessive amounts of glucocorticoids (GCs) are related to the presence and severity of DR, however transcriptional effects of GCs on the biology of human retinal capillary endothelial cells (HRCECs) and its impacts on DR are still unclear. Here, we showed that GC (hydrocortisone) treatment induced ECM component [fibronectin (FN) and type IV collagen (Col IV)] expression and morphological changes in HRCECs via the glucocorticoid receptor (GR), which depended on the nuclear translocation of YAP coactivator. Mechanistically, GCs induced stress fiber formation in HRCECs, while blocking stress fiber formation inhibited GC-induced YAP nuclear translocation. Overexpression of FN, but not Col IV, activated YAP through the promotion of stress fiber formation via ECM-integrin signaling. Thus, a feedforward loop is established to sustain YAP activity. Using mRNA sequencing of HRCECs with overexpressed YAP or GC treatment, we found a similarity in Gene Ontology (GO) terms, differentially expressed genes (DEGs) and transcription factors (TFs) between the two RNA-seq datasets. In vivo, YAP was activated in retina vascular ECs of STZ-induced diabetic mice, and TF prediction analysis of published RNA-seq data of dermal vascular ECs from T2DM patients showed that GR and TEAD (the main transcription factor for YAP) were enriched. Together, GCs activate YAP and promote ECM component (FN and Col IV) remodeling in retinal capillary endothelial cells, and the underlying regulatory mechanism may provide new insights into the vascular BM thickening of the retina in the early pathogenesis of DR.
Collapse
Affiliation(s)
- Xianliang Gu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Lingling Ge
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Bangqi Ren
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yajie Fang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yijian Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Yi Wang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage & Regeneration and Restoration of Chongqing, Chongqing, China
| |
Collapse
|
8
|
Metz JK, Wiegand B, Schnur S, Knoth K, Schneider-Daum N, Groß H, Croston G, Reinheimer TM, Lehr CM, Hittinger M. Modulating the Barrier Function of Human Alveolar Epithelial (hAELVi) Cell Monolayers as a Model of Inflammation. Altern Lab Anim 2021; 48:252-267. [DOI: 10.1177/0261192920983015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The incidence of inflammatory lung diseases such as acute respiratory distress syndrome (ARDS) remains an important problem, particularly in the present time with the Covid-19 pandemic. However, an adequate in vitro test system to monitor the barrier function of the alveolar epithelium during inflammation and for assessing anti-inflammatory drugs is urgently needed. Therefore, we treated human Alveolar Epithelial Lentivirus-immortalised cells (hAELVi cells) with the pro-inflammatory cytokines TNF-α (25 ng/ml) and IFN-γ (30 ng/ml), in the presence or absence of hydrocortisone (HC). While TNF-α and IFN-γ are known to reduce epithelial barrier properties, HC could be expected to protect the barrier function and result in an anti-inflammatory effect. We investigated the impact of anti-inflammatory/inflammatory treatment on transepithelial electrical resistance (TEER) and the apparent permeability coefficient (P app) of the low permeability marker sodium fluorescein (NaFlu). After incubating hAELVi cells for 48 hours with a combination of TNF-α and IFN-γ, there was a significant decrease in TEER and a significant increase in the P app. The presence of HC maintained the TEER values and barrier properties, so that no significant P app change was observed. By using hAELVi cells to study anti-inflammatory drugs in vitro, the need for animal experiments could be reduced and pulmonary drug development accelerated.
Collapse
Affiliation(s)
- Julia Katharina Metz
- PharmBioTec GmbH, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Sabrina Schnur
- PharmBioTec GmbH, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Nicole Schneider-Daum
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | | | | | | | - Claus-Michael Lehr
- Department of Pharmacy, Saarland University, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Marius Hittinger
- PharmBioTec GmbH, Saarbrücken, Germany
- 3RProducts Marius Hittinger, Blieskastel, Germany
| |
Collapse
|
9
|
Latreille E, Lee WL. Interactions of Influenza and SARS-CoV-2 with the Lung Endothelium: Similarities, Differences, and Implications for Therapy. Viruses 2021; 13:161. [PMID: 33499234 PMCID: PMC7911974 DOI: 10.3390/v13020161] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory viruses such as influenza and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are a constant threat to public health given their ability to cause global pandemics. Infection with either virus may lead to aberrant host responses, such as excessive immune cell recruitment and activation, dysregulated inflammation, and coagulopathy. These may contribute to the development of lung edema and respiratory failure. An increasing amount of evidence suggests that lung endothelial cells play a critical role in the pathogenesis of both viruses. In this review, we discuss how infection with influenza or SARS-CoV-2 may induce endothelial dysfunction. We compare the effects of infection of these two viruses, how they may contribute to pathogenesis, and discuss the implications for potential treatment. Understanding the differences between the effects of these two viruses on lung endothelial cells will provide important insight to guide the development of therapeutics.
Collapse
Affiliation(s)
- Elyse Latreille
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Warren L. Lee
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Keenan Centre for Biomedical Research, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Interdepartmental Division of Critical Care and the Department of Medicine, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
10
|
Circulating Von Willebrand factor and high molecular weight multimers as markers of endothelial injury predict COVID-19 in-hospital mortality. Angiogenesis 2021; 24:505-517. [PMID: 33449299 PMCID: PMC7809553 DOI: 10.1007/s10456-020-09762-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022]
Abstract
Background Coronavirus disease 2019 (COVID-19) is a respiratory disease associated with endotheliitis and microthrombosis. Objectives To correlate endothelial dysfunction to in-hospital mortality in a bi-centric cohort of COVID-19 adult patients. Methods Consecutive ambulatory and hospitalized patients with laboratory-confirmed COVID-19 were enrolled. A panel of endothelial biomarkers and von Willebrand factor (VWF) multimers were measured in each patient ≤ 48 h following admission. Results Study enrolled 208 COVID-19 patients of whom 23 were mild outpatients and 189 patients hospitalized after admission. Most of endothelial biomarkers tested were found increased in the 89 critical patients transferred to intensive care unit. However, only von Willebrand factor antigen (VWF:Ag) scaled according to clinical severity, with levels significantly higher in critical patients (median 507%, IQR 428–596) compared to non-critical patients (288%, 230–350, p < 0.0001) or COVID-19 outpatients (144%, 133–198, p = 0.007). Moreover, VWF high molecular weight multimers (HMWM) were significantly higher in critical patients (median ratio 1.18, IQR 0.86–1.09) compared to non-critical patients (0.96, 1.04–1.39, p < 0.001). Among all endothelial biomarkers measured, ROC curve analysis identified a VWF:Ag cut-off of 423% as the best predictor for in-hospital mortality. The accuracy of VWF:Ag was further confirmed in a Kaplan–Meier estimator analysis and a Cox proportional Hazard model adjusted on age, BMI, C-reactive protein and d-dimer levels. Conclusion VWF:Ag is a relevant predictive factor for in-hospital mortality in COVID-19 patients. More than a biomarker, we hypothesize that VWF, including excess of HMWM forms, drives microthrombosis in COVID-19.
Collapse
|
11
|
Espinosa A, Meneses G, Chavarría A, Mancilla R, Pedraza-Chaverri J, Fleury A, Bárcena B, Pérez-Osorio IN, Besedovsky H, Arauz A, Fragoso G, Sciutto E. Intranasal Dexamethasone Reduces Mortality and Brain Damage in a Mouse Experimental Ischemic Stroke Model. Neurotherapeutics 2020; 17:1907-1918. [PMID: 32632775 PMCID: PMC7851226 DOI: 10.1007/s13311-020-00884-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neuroinflammation triggered by the expression of damaged-associated molecular patterns released from dying cells plays a critical role in the pathogenesis of ischemic stroke. However, the benefits from the control of neuroinflammation in the clinical outcome have not been established. In this study, the effectiveness of intranasal, a highly efficient route to reach the central nervous system, and intraperitoneal dexamethasone administration in the treatment of neuroinflammation was evaluated in a 60-min middle cerebral artery occlusion (MCAO) model in C57BL/6 male mice. We performed a side-by-side comparison using intranasal versus intraperitoneal dexamethasone, a timecourse including immediate (0 h) or 4 or 12 h poststroke intranasal administration, as well as 4 intranasal doses of dexamethasone beginning 12 h after the MCAO versus a single dose at 12 h to identify the most effective conditions to treat neuroinflammation in MCAO mice. The best results were obtained 12 h after MCAO and when mice received a single dose of dexamethasone (0.25 mg/kg) intranasally. This treatment significantly reduced mortality, neurological deficits, infarct volume size, blood-brain barrier permeability in the somatosensory cortex, inflammatory cell infiltration, and glial activation. Our results demonstrate that a single low dose of intranasal dexamethasone has neuroprotective therapeutic effects in the MCAO model, showing a better clinical outcome than the intraperitoneal administration. Based on these results, we propose a new therapeutic approach for the treatment of the damage process that accompanies ischemic stroke.
Collapse
Affiliation(s)
- Alejandro Espinosa
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Gabriela Meneses
- Departamento de Parasitología, Instituto Nacional de Diagnóstico y Referencia Epidemiológicos, Mexico City, 01480, Mexico
| | - Anahí Chavarría
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, 06726, Mexico
| | - Raúl Mancilla
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Agnes Fleury
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
- Unidad Periférica del Instituto de Investigaciones Biomédicas en el Instituto Nacional de Neurología y Neurocirugía, Mexico City, 14269, Mexico
| | - Brandon Bárcena
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Ivan N Pérez-Osorio
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Hugo Besedovsky
- The Institute of Physiology and Pathophysiology, Medical Faculty, Philipps University, Marburg, D-35037, Germany
| | - Antonio Arauz
- Stroke Clinic, Instituto Nacional de Neurología y Neurocirugía, Mexico City, 14269, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| | - Edda Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.
| |
Collapse
|
12
|
Zhang F, Zarkada G, Yi S, Eichmann A. Lymphatic Endothelial Cell Junctions: Molecular Regulation in Physiology and Diseases. Front Physiol 2020; 11:509. [PMID: 32547411 PMCID: PMC7274196 DOI: 10.3389/fphys.2020.00509] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Lymphatic endothelial cells (LECs) lining lymphatic vessels develop specialized cell-cell junctions that are crucial for the maintenance of vessel integrity and proper lymphatic vascular functions. Successful lymphatic drainage requires a division of labor between lymphatic capillaries that take up lymph via open "button-like" junctions, and collectors that transport lymph to veins, which have tight "zipper-like" junctions that prevent lymph leakage. In recent years, progress has been made in the understanding of these specialized junctions, as a result of the application of state-of-the-art imaging tools and novel transgenic animal models. In this review, we discuss lymphatic development and mechanisms governing junction remodeling between button and zipper-like states in LECs. Understanding lymphatic junction remodeling is important in order to unravel lymphatic drainage regulation in obesity and inflammatory diseases and may pave the way towards future novel therapeutic interventions.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Georgia Zarkada
- Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Sanjun Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Cardiovascular Research Center, Yale School of Medicine, Yale University, New Haven, CT, United States.,INSERM U970, Paris Cardiovascular Research Center, Paris, France
| |
Collapse
|
13
|
Schroeder T, Bittrich P, Noebel C, Kuhne JF, Schroeder J, Schoen G, Fiehler J, Kniep HC, Gellißen S. Efficiency of Dexamethasone for Treatment of Vasogenic Edema in Brain Metastasis Patients: A Radiographic Approach. Front Oncol 2019; 9:695. [PMID: 31417871 PMCID: PMC6683846 DOI: 10.3389/fonc.2019.00695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/15/2019] [Indexed: 12/27/2022] Open
Abstract
Background and Purpose: To date, imaging studies quantifying the amount of vasogenic edema reduction (VE) in patients with brain metastases (BM) treated with glucocorticoids (GC) have included a very limited number of patients and showed ambiguous results. Here, we aim to determine the radiological effect of GC on VE in BM patients in a large cohort with multiple primary tumor entities in a cross-sectional approach. Materials and Methods: This monocentric retrospective study includes 299 patients first-ever diagnosed with 2,759 intra-axial BM on the respective MRI. 126/299 patients received GC prior to MRI due to mass effect of edema on cranial CT scan and clinical symptoms (GC-pos) and 173 patients did not (GC-neg). GC dose was documented in 85/126 patients. All BM and their respective VE were semi-automatically segmented on post-contrast T1-weighted images. Results: VE volumes were higher in GC-pos compared to GC-neg (p = 0.009) and did not correlate with GC dose. Multivariate linear regression analysis with interaction terms on the assumption that BM volume and BM number influence the probability of GC administration shows that large and higher numbers of BM under GC treatment generate less VE than without (p < 0.001 and p = 0.038, respectively). The primary tumor type and total BM volume did not influence VE volume. Conclusion: Use of GC is especially effective for treatment of VE formation in patients with larger and multiple BM regardless of primary tumor type and dosage. However, based on the present data a direct causative relationship between GC and VE cannot be proven.
Collapse
Affiliation(s)
- Tanja Schroeder
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiology, Schoen Klinik Hamburg Eilbek, Hamburg, Germany
| | - Paul Bittrich
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clara Noebel
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Felix Kuhne
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schroeder
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Schoen
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Fiehler
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helge C Kniep
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Gellißen
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Sato K. [Consideration for future in vitro BBB models - technical development to investigate the drug delivery to the CNS]. Nihon Yakurigaku Zasshi 2019; 152:287-294. [PMID: 30531099 DOI: 10.1254/fpj.152.287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Blood vessels in the central nervous system (CNS) limit the material exchange between blood and parenchyma by blood brain barrier (BBB). At present, no appropriate in vitro BBB models are available for the investigation whether or not the candidate compounds for new drugs could be delivered to the CNS. This causes huge difficulties of the development of CNS drugs and prediction of CNS adverse effects. In this review, I first outline the structures and functions of BBB, together with the parameters used for the quantification of BBB functions. I also introduce the history of in vitro BBB models used in the drug development so far, i.e., the transition from non-cell models to the models using primary culture of rodent cells, porcine, bovine, cell lines, etc. More recently, the application of human cells differentiated from human induced pluripotent stem cells and microfluidic engineering have already started. BBB is essential for the maintenance of brain homeostasis and the mechanisms of the BBB development will be clarified by reproducing functional BBB on the dish. The new in vitro models and the data may provide accurate prediction of drug delivery to the CNS and the improvement of the evaluation system for toxicity and safety, thereby leading to successful launch of new drugs on the market.
Collapse
|
15
|
Wijk A, Canning P, Heijningen RP, Vogels IM, Noorden CJ, Klaassen I, Schlingemann RO. Glucocorticoids exert differential effects on the endothelium in an in vitro model of the blood-retinal barrier. Acta Ophthalmol 2019; 97:214-224. [PMID: 30168271 DOI: 10.1111/aos.13909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/03/2018] [Indexed: 12/25/2022]
Abstract
PURPOSE Glucocorticoids (GCs) are used as treatment in diabetic macular oedema, a condition caused by blood-retinal barrier (BRB) disruption. The proposed mechanisms by which GCs reduce macular oedema are indirect anti-inflammatory effects and inhibition of VEGF production, but direct effects on the BRB endothelium may be equally important. Here, we investigated direct effects of GCs on the endothelium to understand the specific pathways of GC action, to enable development of novel therapeutics lacking the adverse side-effects of the presently used GCs. METHODS Primary bovine retinal endothelial cells (BRECs) were grown on Transwell inserts and treated with hydrocortisone (HC), dexamethasone (Dex) or triamcinolone acetonide (TA). Molecular barrier integrity of the BRB was determined by mRNA and protein expression, and barrier function was assessed using permeability assays. In addition, we investigated whether TA was able to prevent barrier disruption after stimulation with VEGF or cytokines. RESULTS Treatment of BRECs with GCs resulted in upregulation of tight junction mRNA (claudin-5, occludin, ZO-1) and protein (claudin-5 and ZO-1). In functional assays, only TA strengthened the barrier function by reducing endothelial permeability. Moreover, TA was able to prevent cytokine-induced permeability in human retinal endothelial cells and VEGF-induced expression of plasmalemma vesicle-associated protein (PLVAP), a key player in VEGF-induced retinal vascular leakage. CONCLUSION Glucocorticoids have differential effects in an experimental in vitro BRB model. TA is the most potent in improving barrier function, both at the molecular and functional levels, and TA prevents VEGF-induced expression of PLVAP.
Collapse
Affiliation(s)
- Anne‐Eva Wijk
- Departments of Ophthalmology and Medical Biology Amsterdam UMC University of Amsterdam Ocular Angiogenesis Group Amsterdam Cardiovascular Sciences Amsterdam Neuroscience Amsterdam The Netherlands
| | - Paul Canning
- The Wellcome‐Wolfson Institute for Experimental Medicine School of Medicine Dentistry and Biomedical Sciences Queen's University Belfast Belfast UK
| | - Rutger P. Heijningen
- Departments of Ophthalmology and Medical Biology Amsterdam UMC University of Amsterdam Ocular Angiogenesis Group Amsterdam Cardiovascular Sciences Amsterdam Neuroscience Amsterdam The Netherlands
| | - Ilse M.C. Vogels
- Departments of Ophthalmology and Medical Biology Amsterdam UMC University of Amsterdam Ocular Angiogenesis Group Amsterdam Cardiovascular Sciences Amsterdam Neuroscience Amsterdam The Netherlands
| | - Cornelis J.F. Noorden
- Departments of Ophthalmology and Medical Biology Amsterdam UMC University of Amsterdam Ocular Angiogenesis Group Amsterdam Cardiovascular Sciences Amsterdam Neuroscience Amsterdam The Netherlands
- Department of Genetic Toxicology and Cancer Biology National Institute of Biology Ljubljana Slovenia
| | - Ingeborg Klaassen
- Departments of Ophthalmology and Medical Biology Amsterdam UMC University of Amsterdam Ocular Angiogenesis Group Amsterdam Cardiovascular Sciences Amsterdam Neuroscience Amsterdam The Netherlands
| | - Reinier O. Schlingemann
- Departments of Ophthalmology and Medical Biology Amsterdam UMC University of Amsterdam Ocular Angiogenesis Group Amsterdam Cardiovascular Sciences Amsterdam Neuroscience Amsterdam The Netherlands
- Department of Ophthalmology University of Lausanne Jules Gonin Eye Hospital Lausanne Switzerland
| |
Collapse
|
16
|
Keep RF, Andjelkovic AV, Xiang J, Stamatovic SM, Antonetti DA, Hua Y, Xi G. Brain endothelial cell junctions after cerebral hemorrhage: Changes, mechanisms and therapeutic targets. J Cereb Blood Flow Metab 2018; 38:1255-1275. [PMID: 29737222 PMCID: PMC6092767 DOI: 10.1177/0271678x18774666] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 11/15/2022]
Abstract
Vascular disruption is the underlying cause of cerebral hemorrhage, including intracerebral, subarachnoid and intraventricular hemorrhage. The disease etiology also involves cerebral hemorrhage-induced blood-brain barrier (BBB) disruption, which contributes an important component to brain injury after the initial cerebral hemorrhage. BBB loss drives vasogenic edema, allows leukocyte extravasation and may lead to the entry of potentially neurotoxic and vasoactive compounds into brain. This review summarizes current information on changes in brain endothelial junction proteins in response to cerebral hemorrhage (and clot-related factors), the mechanisms underlying junction modification and potential therapeutic targets to limit BBB disruption and, potentially, hemorrhage occurrence. It also addresses advances in the tools that are now available for assessing changes in junctions after cerebral hemorrhage and the potential importance of such junction changes. Recent studies suggest post-translational modification, conformational change and intracellular trafficking of junctional proteins may alter barrier properties. Understanding how cerebral hemorrhage alters BBB properties beyond changes in tight junction protein loss may provide important therapeutic insights to prevent BBB dysfunction and restore normal function.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Ann Arbor, MI, USA
| | - Jianming Xiang
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| | | | - David A Antonetti
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
- Department of Ophthalmology & Visual Science Medical School, University of Michigan Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
17
|
Hesam Mahmoudinezhad M, Karkhaneh A, Jadidi K. Effect of PEDOT:PSS in tissue engineering composite scaffold on improvement and maintenance of endothelial cell function. J Biosci 2018. [DOI: 10.1007/s12038-018-9748-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
18
|
Juszczak GR, Stankiewicz AM. Glucocorticoids, genes and brain function. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:136-168. [PMID: 29180230 DOI: 10.1016/j.pnpbp.2017.11.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 10/18/2017] [Accepted: 11/23/2017] [Indexed: 01/02/2023]
Abstract
The identification of key genes in transcriptomic data constitutes a huge challenge. Our review of microarray reports revealed 88 genes whose transcription is consistently regulated by glucocorticoids (GCs), such as cortisol, corticosterone and dexamethasone, in the brain. Replicable transcriptomic data were combined with biochemical and physiological data to create an integrated view of the effects induced by GCs. The most frequently reported genes were Errfi1 and Ddit4. Their up-regulation was associated with the altered transcription of genes regulating growth factor and mTORC1 signaling (Gab1, Tsc22d3, Dusp1, Ndrg2, Ppp5c and Sesn1) and progression of the cell cycle (Ccnd1, Cdkn1a and Cables1). The GC-induced reprogramming of cell function involves changes in the mRNA level of genes responsible for the regulation of transcription (Klf9, Bcl6, Klf15, Tle3, Cxxc5, Litaf, Tle4, Jun, Sox4, Sox2, Sox9, Irf1, Sall2, Nfkbia and Id1) and the selective degradation of mRNA (Tob2). Other genes are involved in the regulation of metabolism (Gpd1, Aldoc and Pdk4), actin cytoskeleton (Myh2, Nedd9, Mical2, Rhou, Arl4d, Osbpl3, Arhgef3, Sdc4, Rdx, Wipf3, Chst1 and Hepacam), autophagy (Eva1a and Plekhf1), vesicular transport (Rhob, Ehd3, Vps37b and Scamp2), gap junctions (Gjb6), immune response (Tiparp, Mertk, Lyve1 and Il6r), signaling mediated by thyroid hormones (Thra and Sult1a1), calcium (Calm2), adrenaline/noradrenaline (Adcy9 and Adra1d), neuropeptide Y (Npy1r) and histamine (Hdc). GCs also affected genes involved in the synthesis of polyamines (Azin1) and taurine (Cdo1). The actions of GCs are restrained by feedback mechanisms depending on the transcription of Sgk1, Fkbp5 and Nr3c1. A side effect induced by GCs is increased production of reactive oxygen species. Available data show that the brain's response to GCs is part of an emergency mode characterized by inactivation of non-core activities, restrained inflammation, restriction of investments (growth), improved efficiency of energy production and the removal of unnecessary or malfunctioning cellular components to conserve energy and maintain nutrient supply during the stress response.
Collapse
Affiliation(s)
- Grzegorz R Juszczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland.
| | - Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552 Magdalenka, Poland
| |
Collapse
|
19
|
Interleukin 6-Mediated Endothelial Barrier Disturbances Can Be Attenuated by Blockade of the IL6 Receptor Expressed in Brain Microvascular Endothelial Cells. Transl Stroke Res 2018; 9:631-642. [PMID: 29429002 DOI: 10.1007/s12975-018-0614-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 01/01/2023]
Abstract
Compromised blood-brain barrier (BBB) by dysregulation of cellular junctions is a hallmark of many cerebrovascular disorders due to the pro-inflammatory cytokines action. Interleukin 6 (IL6) is implicated in inflammatory processes and in secondary brain injury after subarachnoid hemorrhage (SAH) but its role in the maintenance of cerebral endothelium still requires a precise elucidation. Although IL6 has been shown to exert pro-inflammatory action on brain microvascular endothelial cells (ECs), the expression of one of the IL6 receptors, the IL6R is controversially discussed. In attempt to reach more clarity in this issue, we present here an evident baseline expression of the IL6R in BBB endothelium in vivo and in an in vitro model of the BBB, the cEND cell line. A significantly increased expression of IL6R and its ligand was observed in BBB capillaries 2 days after experimental SAH in mice. In vitro, we saw IL6 administration resulting in an intracellular and extracellular elevation of IL6 protein, which was accompanied by a reduced expression of tight and adherens junctions, claudin-5, occludin, and vascular-endothelial (VE-) cadherin. By functional assays, we could demonstrate IL6-incubated brain ECs to lose their endothelial integrity that can be attenuated by inhibiting the IL6R. Blockade of the IL6R by a neutralizing antibody has reconstituted the intercellular junction expression to the control level and caused a restoration of the transendothelial electrical resistance of the cEND cell monolayer. Our findings add depth to the current understanding of the involvement of the endothelial IL6R in the loss of EC integrity implicating potential therapy options.
Collapse
|
20
|
Na W, Shin JY, Lee JY, Jeong S, Kim WS, Yune TY, Ju BG. Dexamethasone suppresses JMJD3 gene activation via a putative negative glucocorticoid response element and maintains integrity of tight junctions in brain microvascular endothelial cells. J Cereb Blood Flow Metab 2017; 37:3695-3708. [PMID: 28338398 PMCID: PMC5718327 DOI: 10.1177/0271678x17701156] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB) exhibits a highly selective permeability to support the homeostasis of the central nervous system (CNS). The tight junctions in the BBB microvascular endothelial cells seal the paracellular space to prevent diffusion. Thus, disruption of tight junctions results in harmful effects in CNS diseases and injuries. It has recently been demonstrated that glucocorticoids have beneficial effects on maintaining tight junctions in both in vitro cell and in vivo animal models. In the present study, we found that dexamethasone suppresses the expression of JMJD3, a histone H3K27 demethylase, via the recruitment of glucocorticoid receptor α (GRα) and nuclear receptor co-repressor (N-CoR) to the negative glucocorticoid response element (nGRE) in the upstream region of JMJD3 gene in brain microvascular endothelial cells subjected to TNFα treatment. The decreased JMJD3 gene expression resulted in the suppression of MMP-2, MMP-3, and MMP-9 gene activation. Dexamethasone also activated the expression of the claudin 5 and occludin genes. Collectively, dexamethasone attenuated the disruption of the tight junctions in the brain microvascular endothelial cells subjected to TNFα treatment. Therefore, glucocorticoids may help to preserve the integrity of the tight junctions in the BBB via transcriptional and post-translational regulation following CNS diseases and injuries.
Collapse
Affiliation(s)
- Wonho Na
- 1 Department of Life Science, Sogang University, Seoul, Korea
| | - Jee Y Shin
- 1 Department of Life Science, Sogang University, Seoul, Korea
| | - Jee Y Lee
- 2 Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul, Korea
| | - Sangyun Jeong
- 3 Department of Molecular Biology, Chonbuk National University, Jeonju, Korea
| | - Won-Sun Kim
- 1 Department of Life Science, Sogang University, Seoul, Korea
| | - Tae Y Yune
- 2 Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul, Korea.,4 Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Bong-Gun Ju
- 1 Department of Life Science, Sogang University, Seoul, Korea
| |
Collapse
|
21
|
Dang TQ, Yoon N, Chasiotis H, Dunford EC, Feng Q, He P, Riddell MC, Kelly SP, Sweeney G. Transendothelial movement of adiponectin is restricted by glucocorticoids. J Endocrinol 2017; 234:101-114. [PMID: 28705835 PMCID: PMC6231241 DOI: 10.1530/joe-16-0363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/10/2017] [Indexed: 12/31/2022]
Abstract
Altered permeability of the endothelial barrier in a variety of tissues has implications both in disease pathogenesis and treatment. Glucocorticoids are potent mediators of endothelial permeability, and this forms the basis for their heavily prescribed use as medications to treat ocular disease. However, the effect of glucocorticoids on endothelial barriers elsewhere in the body is less well studied. Here, we investigated glucocorticoid-mediated changes in endothelial flux of Adiponectin (Ad), a hormone with a critical role in diabetes. First, we used monolayers of endothelial cells in vitro and found that the glucocorticoid dexamethasone increased transendothelial electrical resistance and reduced permeability of polyethylene glycol (PEG, molecular weight 4000 Da). Dexamethasone reduced flux of Ad from the apical to basolateral side, measured both by ELISA and Western blotting. We then examined a diabetic rat model induced by treatment with exogenous corticosterone, which was characterized by glucose intolerance and hyperinsulinemia. There was no change in circulating Ad but less Ad protein in skeletal muscle homogenates, despite slightly higher mRNA levels, in diabetic vs control muscles. Dexamethasone-induced changes in Ad flux across endothelial monolayers were associated with alterations in the abundance of select claudin tight junction (TJ) proteins. shRNA-mediated knockdown of one such gene, claudin-7, in HUVEC resulted in decreased TEER and increased adiponectin flux, confirming the functional significance of Dex-induced changes in its expression. In conclusion, our study identifies glucocorticoid-mediated reductions in flux of Ad across endothelial monolayers in vivo and in vitro This suggests that impaired Ad action in target tissues, as a consequence of reduced transendothelial flux, may contribute to the glucocorticoid-induced diabetic phenotype.
Collapse
Affiliation(s)
- Thanh Q Dang
- Department of BiologyFaculty of Science York University, Toronto, Canada
| | - Nanyoung Yoon
- Department of BiologyFaculty of Science York University, Toronto, Canada
| | - Helen Chasiotis
- Department of BiologyFaculty of Science York University, Toronto, Canada
| | - Emily C Dunford
- School of Kinesiology and Health ScienceFaculty of Health and Muscle Health Research Center, York University, Toronto, Canada
| | - Qilong Feng
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Pingnian He
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA, USA
| | - Michael C Riddell
- School of Kinesiology and Health ScienceFaculty of Health and Muscle Health Research Center, York University, Toronto, Canada
| | - Scott P Kelly
- Department of BiologyFaculty of Science York University, Toronto, Canada
| | - Gary Sweeney
- Department of BiologyFaculty of Science York University, Toronto, Canada
| |
Collapse
|
22
|
Blecharz KG, Frey D, Schenkel T, Prinz V, Bedini G, Krug SM, Czabanka M, Wagner J, Fromm M, Bersano A, Vajkoczy P. Autocrine release of angiopoietin-2 mediates cerebrovascular disintegration in Moyamoya disease. J Cereb Blood Flow Metab 2017; 37:1527-1539. [PMID: 27381827 PMCID: PMC5453470 DOI: 10.1177/0271678x16658301] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Moyamoya disease is a rare steno-occlusive cerebrovascular disorder often resulting in hemorrhagic and ischemic strokes. Although sharing the same ischemic stimulus with atherosclerotic cerebrovascular disease, Moyamoya disease is characterized by a highly instable cerebrovascular system which is prone to rupture due to pathological neovascularization. To understand the molecular mechanisms underlying this instability, angiopoietin-2 gene expression was analyzed in middle cerebral artery lesions obtained from Moyamoya disease and atherosclerotic cerebrovascular disease patients. Angiopoietin-2 was significantly up-regulated in Moyamoya vessels, while serum concentrations of soluble angiopoietins were not changed. For further evaluations, cerebral endothelial cells incubated with serum from these patients in vitro were applied. In contrast to atherosclerotic cerebrovascular disease serum, Moyamoya disease serum induced an angiopoietin-2 overexpression and secretion, accompanied by loss of endothelial integrity. These effects were absent or inverse in endothelial cells of non-brain origin suggesting brain endothelium specificity. The destabilizing effects on brain endothelial cells to Moyamoya disease serum were partially suppressed by the inhibition of angiopoietin-2. Our findings define brain endothelial cells as the potential source of vessel-destabilizing factors inducing the high plasticity state and disintegration in Moyamoya disease in an autocrine manner. We also provide new insights into Moyamoya disease pathophysiology that may be helpful for preventive treatment strategies in future.
Collapse
Affiliation(s)
- Kinga G Blecharz
- 1 Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Frey
- 2 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Schenkel
- 1 Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Vincent Prinz
- 2 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gloria Bedini
- 3 Laboratory of Cellular Neurobiology, Neurology Unit, UCV, Milan, Italy.,4 Neurological Institute "C. Besta", Milan, Italy
| | - Susanne M Krug
- 5 Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Czabanka
- 2 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Josephin Wagner
- 1 Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Fromm
- 5 Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Bersano
- 6 Neurology Unit, UCV, Milan, Italy.,7 Neurological Institute "C. Besta", IRCCS Foundation, Milan, Italy
| | - Peter Vajkoczy
- 1 Department of Experimental Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,2 Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany.,8 Center of Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
23
|
Hu X, De Silva TM, Chen J, Faraci FM. Cerebral Vascular Disease and Neurovascular Injury in Ischemic Stroke. Circ Res 2017; 120:449-471. [PMID: 28154097 PMCID: PMC5313039 DOI: 10.1161/circresaha.116.308427] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 12/13/2022]
Abstract
The consequences of cerebrovascular disease are among the leading health issues worldwide. Large and small cerebral vessel disease can trigger stroke and contribute to the vascular component of other forms of neurological dysfunction and degeneration. Both forms of vascular disease are driven by diverse risk factors, with hypertension as the leading contributor. Despite the importance of neurovascular disease and subsequent injury after ischemic events, fundamental knowledge in these areas lag behind our current understanding of neuroprotection and vascular biology in general. The goal of this review is to address select key structural and functional changes in the vasculature that promote hypoperfusion and ischemia, while also affecting the extent of injury and effectiveness of therapy. In addition, as damage to the blood-brain barrier is one of the major consequences of ischemia, we discuss cellular and molecular mechanisms underlying ischemia-induced changes in blood-brain barrier integrity and function, including alterations in endothelial cells and the contribution of pericytes, immune cells, and matrix metalloproteinases. Identification of cell types, pathways, and molecules that control vascular changes before and after ischemia may result in novel approaches to slow the progression of cerebrovascular disease and lessen both the frequency and impact of ischemic events.
Collapse
Affiliation(s)
- Xiaoming Hu
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - T. Michael De Silva
- Biomedicine Discovery Institute, Department of Pharmacology, 9 Ancora Imparo Way, Monash University, Clayton, Vic, Australia
| | - Jun Chen
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213
| | - Frank M. Faraci
- Departments of Internal Medicine and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City Veterans Affairs Healthcare System, Iowa City, IA, USA
| |
Collapse
|
24
|
Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE. Endothelial Response to Glucocorticoids in Inflammatory Diseases. Front Immunol 2016; 7:592. [PMID: 28018358 PMCID: PMC5155119 DOI: 10.3389/fimmu.2016.00592] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022] Open
Abstract
The endothelium plays a crucial role in inflammation. A balanced control of inflammation requires the action of glucocorticoids (GCs), steroidal hormones with potent cell-specific anti-inflammatory properties. Besides the classic anti-inflammatory effects of GCs on leukocytes, recent studies confirm that endothelial cells also represent an important target for GCs. GCs regulate different aspects of endothelial physiology including expression of adhesion molecules, production of pro-inflammatory cytokines and chemokines, and maintenance of endothelial barrier integrity. However, the regulation of endothelial GC sensitivity remains incompletely understood. In this review, we specifically examine the endothelial response to GCs in various inflammatory diseases ranging from multiple sclerosis, stroke, sepsis, and vasculitis to atherosclerosis. Shedding more light on the cross talk between GCs and endothelium will help to improve existing therapeutic strategies and develop new therapies better tailored to the needs of patients.
Collapse
Affiliation(s)
- Karolina A. Zielińska
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Laura Van Moortel
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent, VIB Medical Biotechnology Center, Ghent, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Karolien De Bosscher
- Receptor Research Laboratories, Nuclear Receptor Lab, VIB-UGent, VIB Medical Biotechnology Center, Ghent, Belgium
| | | |
Collapse
|
25
|
Murayi R, Chittiboina P. Glucocorticoids in the management of peritumoral brain edema: a review of molecular mechanisms. Childs Nerv Syst 2016; 32:2293-2302. [PMID: 27613642 PMCID: PMC5136308 DOI: 10.1007/s00381-016-3240-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 08/31/2016] [Indexed: 12/15/2022]
Abstract
Peritumoral brain edema (PTBE) is mediated by blood-brain barrier breakdown. PTBE results from interstitial vasogenic brain edema due to vascular endothelial growth factor and other inflammatory products of brain tumors. Glucocorticoids (GCs) are the mainstay for treatment of PTBE despite significant systemic side effects. GCs are thought to affect multiple cell types in the edematous brain. Here, we review preclinical studies of GC effects on edematous brain and review mechanisms underlying GC action on tumor cells, endothelial cells, and astrocytes. GCs may reduce tumor cell viability and suppress vascular endothelial growth factor (VEGF) production in tumor cells. Modulation of expression and distribution of tight junction proteins occludin, claudin-5, and ZO-1 in endothelial cells likely plays a central role in GC action on endothelial cells. GCs may also have an effect on astrocyte angiopoietin production and limited effect on astrocyte aquaporin. A better understanding of these molecular mechanisms may lead to the development of novel therapeutics for management of PTBE with a better side effect profile.
Collapse
Affiliation(s)
- Roger Murayi
- Surgical Neurology Branch, Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Diseases and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD, 20892-1414, USA
| | - Prashant Chittiboina
- Surgical Neurology Branch, Neurosurgery Unit for Pituitary and Inheritable Diseases, National Institute of Neurological Diseases and Stroke, National Institutes of Health, 10 Center Drive, Room 3D20, Bethesda, MD, 20892-1414, USA.
| |
Collapse
|
26
|
Salvador E, Burek M, Förster CY. Tight Junctions and the Tumor Microenvironment. CURRENT PATHOBIOLOGY REPORTS 2016; 4:135-145. [PMID: 27547510 PMCID: PMC4978755 DOI: 10.1007/s40139-016-0106-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Tight junctions (TJs) are specialized differentiations of epithelial and endothelial cell membranes. TJs play an important role in the adhesion of cells and their interaction with each other. Most cancers originate from epithelial cells. Thus, it is of significance to examine the role of TJs in the tumor microenvironment (TME) and how they affect cancer metastasis. RECENT FINDINGS In epithelium-derived cancers, intactness of the primary tumor mass is influenced by intercellular structures as well as cell-to-cell adhesion. Irregularities of these factors may lead to tumor dissociation and subsequent metastasis. Low expression of TJs is observed among highly metastatic cancer cells. SUMMARY In this review, we summarized findings from current literature in consideration of the role of TJs in relation to the TME and cancer. Deeper understanding of the mechanisms leading to TJ dysregulation is needed to facilitate the design and conceptualization of new and better therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Ellaine Salvador
- Department of Anesthesia and Critical Care, University of Wurzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Malgorzata Burek
- Department of Anesthesia and Critical Care, University of Wurzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Carola Y. Förster
- Department of Anesthesia and Critical Care, University of Wurzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| |
Collapse
|
27
|
Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Förster C, Galla HJ, Romero IA, Shusta EV, Stebbins MJ, Vandenhaute E, Weksler B, Brodin B. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab 2016; 36:862-90. [PMID: 26868179 PMCID: PMC4853841 DOI: 10.1177/0271678x16630991] [Citation(s) in RCA: 525] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
The endothelial cells lining the brain capillaries separate the blood from the brain parenchyma. The endothelial monolayer of the brain capillaries serves both as a crucial interface for exchange of nutrients, gases, and metabolites between blood and brain, and as a barrier for neurotoxic components of plasma and xenobiotics. This "blood-brain barrier" function is a major hindrance for drug uptake into the brain parenchyma. Cell culture models, based on either primary cells or immortalized brain endothelial cell lines, have been developed, in order to facilitate in vitro studies of drug transport to the brain and studies of endothelial cell biology and pathophysiology. In this review, we aim to give an overview of established in vitro blood-brain barrier models with a focus on their validation regarding a set of well-established blood-brain barrier characteristics. As an ideal cell culture model of the blood-brain barrier is yet to be developed, we also aim to give an overview of the advantages and drawbacks of the different models described.
Collapse
Affiliation(s)
- Hans C Helms
- Department of Pharmacy, University of Copenhagen, Denmark
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, UK
| | - Malgorzata Burek
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | | | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, HAS, Szeged, Hungary
| | - Carola Förster
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | - Hans J Galla
- Institute of Biochemistry, University of Muenster, Germany
| | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | | | - Babette Weksler
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, NY, USA
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Denmark
| |
Collapse
|
28
|
Blecharz KG, Colla R, Rohde V, Vajkoczy P. Control of the blood-brain barrier function in cancer cell metastasis. Biol Cell 2015; 107:342-71. [PMID: 26032862 DOI: 10.1111/boc.201500011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/22/2015] [Indexed: 12/25/2022]
Abstract
Cerebral metastases are the most common brain neoplasms seen clinically in the adults and comprise more than half of all brain tumours. Actual treatment options for brain metastases that include surgical resection, radiotherapy and chemotherapy are rarely curative, although palliative treatment improves survival and life quality of patients carrying brain-metastatic tumours. Chemotherapy in particular has also shown limited or no activity in brain metastasis of most tumour types. Many chemotherapeutic agents used systemically do not cross the blood-brain barrier (BBB), whereas others may transiently weaken the BBB and allow extravasation of tumour cells from the circulation into the brain parenchyma. Increasing evidence points out that the interaction between the BBB and tumour cells plays a key role for implantation and growth of brain metastases in the central nervous system. The BBB, as the tightest endothelial barrier, prevents both early detection and treatment by creating a privileged microenvironment. Therefore, as observed in several in vivo studies, precise targetting the BBB by a specific transient opening of the structure making it permeable for therapeutic compounds, might potentially help to overcome this difficult clinical problem. Moreover, a better understanding of the molecular features of the BBB, its interrelation with metastatic tumour cells and the elucidation of cellular mechanisms responsible for establishing cerebral metastasis must be clearly outlined in order to promote treatment modalities that particularly involve chemotherapy. This in turn would substantially expand the survival and quality of life of patients with brain metastasis, and potentially increase the remission rate. Therefore, the focus of this review is to summarise the current knowledge on the role and function of the BBB in cancer metastasis.
Collapse
Affiliation(s)
- Kinga G Blecharz
- Department of Experimental Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, 10119, Germany
| | - Ruben Colla
- Department of Neurosurgery, Göttingen University Medical Center, Göttingen, 37070, Germany
| | - Veit Rohde
- Department of Neurosurgery, Göttingen University Medical Center, Göttingen, 37070, Germany
| | - Peter Vajkoczy
- Department of Experimental Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, 10119, Germany.,Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, 13353, Germany
| |
Collapse
|
29
|
Hydrocortisone enhances the barrier properties of HBMEC/ciβ, a brain microvascular endothelial cell line, through mesenchymal-to-endothelial transition-like effects. Fluids Barriers CNS 2015; 12:7. [PMID: 25763180 PMCID: PMC4355132 DOI: 10.1186/s12987-015-0003-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/22/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Because in vitro blood-brain barrier (BBB) models are important tools for studying brain diseases and drug development, we recently established a new line of conditionally immortalized human brain microvascular endothelial cells (HBMEC/ciβ) for use in such models. Since one of the most important functional features of the BBB is its strong intercellular adhesion, in this study, we aimed at improving HBMEC/ciβ barrier properties by means of culture media modifications, thus enhancing their use for future BBB studies. In addition, we simultaneously attempted to obtain insights on related mechanistic properties. METHODS Several types of culture media were prepared in an effort to identify the medium most suitable for culturing HBMEC/ciβ. The barrier properties of HBMEC/ciβ were examined by determining Na(+)-fluorescein permeability and transendothelial electric resistance (TEER). Endothelial marker mRNA expression levels were determined by quantitative real-time polymerase chain reaction. Adherens junction (AJ) formation was examined by immunocytochemistry. Cell migration ability was analyzed by scratch assay. Furthermore, cellular lipid composition was examined by liquid chromatography-time-of-flight mass spectrometry. RESULTS Our initial screening tests showed that addition of hydrocortisone (HC) to the basal medium significantly reduced the Na(+)-fluorescein permeability and increased the TEER of HBMEC/ciβ monolayers. It was also found that, while AJ proteins were diffused in the cytoplasm of HBMEC/ciβ cultured without HC, those expressed in cells cultured with HC were primarily localized at the cell border. Furthermore, this facilitation of AJ formation by HC was in concert with increased endothelial marker mRNA levels and increased ether-type phosphatidylethanolamine levels, while cell migration was retarded in the presence of HC. CONCLUSIONS Our results show that HC supplementation to the basal medium significantly enhances the barrier properties of HBMEC/ciβ. This was associated with a marked phenotypic alteration in HBMEC/ciβ through orchestration of various signaling pathways. Taken together, it appears that overall effects of HC on HBMEC/ciβ could be summarized as facilitating endothelial differentiation characteristics while concurrently retarding mesenchymal characteristics.
Collapse
|
30
|
Thal SC, Neuhaus W. The blood-brain barrier as a target in traumatic brain injury treatment. Arch Med Res 2014; 45:698-710. [PMID: 25446615 DOI: 10.1016/j.arcmed.2014.11.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) is one of the most frequent causes of death in the young population. Several clinical trials have unsuccessfully focused on direct neuroprotective therapies. Recently immunotherapeutic strategies shifted into focus of translational research in acute CNS diseases. Cross-talk between activated microglia and blood-brain barrier (BBB) could initiate opening of the BBB and subsequent recruitment of systemic immune cells and mediators into the brain. Stabilization of the BBB after TBI could be a promising strategy to limit neuronal inflammation, secondary brain damage and acute neurodegeneration. This review provides an overview on the pathophysiology of TBI and brain edema formation including definitions and classification of TBI, current clinical treatment strategies, as well as current understanding on the underlying cellular processes. A summary of in vivo and in vitro models to study different aspects of TBI is presented. Three mechanisms proposed for stabilization of the BBB, myosin light chain kinases, glucocorticoid receptors and peroxisome proliferator-activated receptors are reviewed for their influence on barrier-integrity and outcome after TBI. In conclusion, the BBB is recommended as a promising target for the treatment of traumatic brain injury, and it is suggested that a combination of BBB stabilization and neuroprotectants may improve therapeutic success.
Collapse
Affiliation(s)
- Serge C Thal
- Department of Anesthesia and Critical Care, Johannes Gutenberg University, Mainz, Germany
| | - Winfried Neuhaus
- Department of Pharmaceutical Chemistry, University of Vienna, Althanstrasse, Vienna, Austria; Department of Anesthesia and Critical Care, University Hospital Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
31
|
Firestone GL, Kapadia BJ. Minireview: Steroid/nuclear receptor-regulated dynamics of occluding and anchoring junctions. Mol Endocrinol 2014; 28:1769-84. [PMID: 25203673 DOI: 10.1210/me.2014-1037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A diverse set of physiological signals control intercellular interactions by regulating the structure and function of occluding junctions (tight junctions) and anchoring junctions (adherens junctions and desmosomes). These plasma membrane junctions are comprised of multiprotein complexes of transmembrane and cytoplasmic peripheral plasma membrane proteins. Evidence from many hormone-responsive tissues has shown that expression, modification, molecular interactions, stability, and localization of junctional complex-associated proteins can be targeted by nuclear hormone receptors and their ligands through transcriptional and nontranscriptional mechanisms. The focus of this minireview is to discuss molecular, cellular, and physiological studies that directly link nuclear receptor- and ligand-triggered signaling pathways to the regulation of occluding and anchoring junction dynamics.
Collapse
Affiliation(s)
- Gary L Firestone
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, University of California at Berkeley, Berkeley, California 94720-3200
| | | |
Collapse
|
32
|
García-Ponce A, Citalán-Madrid AF, Velázquez-Avila M, Vargas-Robles H, Schnoor M. The role of actin-binding proteins in the control of endothelial barrier integrity. Thromb Haemost 2014; 113:20-36. [PMID: 25183310 DOI: 10.1160/th14-04-0298] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/01/2014] [Indexed: 01/19/2023]
Abstract
The endothelial barrier of the vasculature is of utmost importance for separating the blood stream from underlying tissues. This barrier is formed by tight and adherens junctions (TJ and AJ) that form intercellular endothelial contacts. TJ and AJ are integral membrane structures that are connected to the actin cytoskeleton via various adaptor molecules. Consequently, the actin cytoskeleton plays a crucial role in regulating the stability of endothelial cell contacts and vascular permeability. While a circumferential cortical actin ring stabilises junctions, the formation of contractile stress fibres, e. g. under inflammatory conditions, can contribute to junction destabilisation. However, the role of actin-binding proteins (ABP) in the control of vascular permeability has long been underestimated. Naturally, ABP regulate permeability via regulation of actin remodelling but some actin-binding molecules can also act independently of actin and control vascular permeability via various signalling mechanisms such as activation of small GTPases. Several studies have recently been published highlighting the importance of actin-binding molecules such as cortactin, ezrin/radixin/moesin, Arp2/3, VASP or WASP for the control of vascular permeability by various mechanisms. These proteins have been described to regulate vascular permeability under various pathophysiological conditions and are thus of clinical relevance as targets for the development of treatment strategies for disorders that are characterised by vascular hyperpermeability such as sepsis. This review highlights recent advances in determining the role of ABP in the control of endothelial cell contacts and vascular permeability.
Collapse
Affiliation(s)
| | | | | | | | - Michael Schnoor
- Dr. Michael Schnoor, CINVESTAV del IPN, Department for Molecular Biomedicine, Av. IPN 2508, San Pedro Zacatenco, GAM, 07360 Mexico City, Mexico, Tel.: +52 55 5747 3321, Fax: +52 55 5747 3938, E-mail:
| |
Collapse
|
33
|
Witt KA, Sandoval KE. Steroids and the blood-brain barrier: therapeutic implications. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:361-390. [PMID: 25307223 DOI: 10.1016/bs.apha.2014.06.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Steroids have a wide spectrum of impact, serving as fundamental regulators of nearly every physiological process within the human body. Therapeutic applications of steroids are equally broad, with a diverse range of medications and targets. Within the central nervous system (CNS), steroids influence development, memory, behavior, and disease outcomes. Moreover, steroids are well recognized as to their impact on the vascular endothelium. The blood-brain barrier (BBB) at the level of the brain microvascular endothelium serves as the principle interface between the peripheral circulation and the brain. Steroids have been identified to impact several critical properties of the BBB, including cellular efflux mechanisms, nutrient uptake, and tight junction integrity. Such actions not only influence brain homeostasis but also the delivery of CNS-targeted therapeutics. A greater understanding of the respective steroid-BBB interactions may shed further light on the differential treatment outcomes observed across CNS pathologies. In this chapter, we examine the current therapeutic implications of steroids respective to BBB structure and function, with emphasis on glucocorticoids and estrogens.
Collapse
Affiliation(s)
- Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University, Edwardsville, Illinois, USA.
| | - Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University, Edwardsville, Illinois, USA
| |
Collapse
|
34
|
Blecharz KG, Burek M, Bauersachs J, Thum T, Tsikas D, Widder J, Roewer N, Förster CY. Inhibition of proteasome-mediated glucocorticoid receptor degradation restores nitric oxide bioavailability in myocardial endothelial cells in vitro. Biol Cell 2014; 106:219-35. [PMID: 24749543 DOI: 10.1111/boc.201300083] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 04/15/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND INFORMATION Glucocorticoids (GCs), including the synthetic GC derivate dexamethasone, are widely used as immunomodulators. One of the numerous side effects of dexamethasone therapy is hypertension arising from reduced release of the endothelium-derived vasodilator nitric oxide (NO). RESULTS Herein, we described the role of dexamethasone and its glucocorticoid receptor (GR) in the regulation of NO synthesis in vitro using the mouse myocardial microvascular endothelial cell line, MyEND. GC treatment caused a firm decrease of extracellular NO levels, whereas the expression of endothelial NO synthase (eNOS) was not affected. However, GC application induced an impairment of tetrahydrobiopterin (BH4 ) concentrations as well as GTP cyclohydrolase-1 (GTPCH-1) expression, both essential factors for NO production upstream of eNOS. Moreover, dexamethasone stimulation resulted in a substantially decreased GR gene and protein expression in MyEND cells. Importantly, inhibition of proteasome-mediated proteolysis of the GR or overexpression of an ubiquitination-defective GR construct improved the bioavailability of BH4 and strengthened GTPCH-1 expression and eNOS activity. CONCLUSIONS Summarising our results, we propose a new mechanism involved in the regulation of NO signalling by GCs in myocardial endothelial cells. We suggest that a sufficient GR protein expression plays a crucial role for the management of GC-induced harmful adverse effects, including deregulations of vasorelaxation arising from disturbed NO biosynthesis.
Collapse
Affiliation(s)
- Kinga G Blecharz
- University of Würzburg, Department of Anaesthesia and Critical Care, Würzburg, 97080, Germany
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Glucocorticoids and endothelial cell barrier function. Cell Tissue Res 2013; 355:597-605. [PMID: 24352805 PMCID: PMC3972429 DOI: 10.1007/s00441-013-1762-z] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/05/2013] [Indexed: 01/14/2023]
Abstract
Glucocorticoids (GCs) are steroid hormones that have inflammatory and immunosuppressive effects on a wide variety of cells. They are used as therapy for inflammatory disease and as a common agent against edema. The blood brain barrier (BBB), comprising microvascular endothelial cells, serves as a permeability screen between the blood and the brain. As such, it maintains homeostasis of the central nervous system (CNS). In many CNS disorders, BBB integrity is compromised. GC treatment has been demonstrated to improve the tightness of the BBB. The responses and effects of GCs are mediated by the ubiquitous GC receptor (GR). Ligand-bound GR recognizes and binds to the GC response element located within the promoter region of target genes. Transactivation of certain target genes leads to improved barrier properties of endothelial cells. In this review, we deal with the role of GCs in endothelial cell barrier function. First, we describe the mechanisms of GC action at the molecular level. Next, we discuss the regulation of the BBB by GCs, with emphasis on genes targeted by GCs such as occludin, claudins and VE-cadherin. Finally, we present currently available GC therapeutic strategies and their limitations.
Collapse
|
36
|
Inhibition of proteasomal glucocorticoid receptor degradation restores dexamethasone-mediated stabilization of the blood-brain barrier after traumatic brain injury. Crit Care Med 2013; 41:1305-15. [PMID: 23474678 DOI: 10.1097/ccm.0b013e31827ca494] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To establish the molecular background for glucocorticoid insensitivity, that is, failure to reduce edema formation and to protect blood-brain barrier integrity after acute traumatic brain injury. DESIGN Controlled animal study. SETTING University research laboratory. SUBJECTS Male C57Bl/6N mice. INTERVENTIONS Mechanical brain lesion by controlled cortical impact. MEASUREMENTS AND MAIN RESULTS Our study demonstrates that 1) proteasomal glucocorticoid receptor degradation is established in brain endothelial cells after traumatic brain injury as a form of posttranslational glucocorticoid receptor modification; 2) inhibition of the proteasomal degradation pathway with bortezomib (0.2 mg/kg) in combination with the glucocorticoid dexamethasone (10 mg/kg) by subcutaneous injection 30 minutes postinjury restores levels of barrier sealing glucocorticoid receptor target occludin in brain endothelial cells, improves blood-brain barrier integrity, reduces edema formation, and limits neuronal damage after brain trauma. CONCLUSIONS The results indicate that the stabilizing effect of glucocorticoids on the blood-brain barrier is hampered after cerebral lesions by proteasomal glucocorticoid receptor degradation in brain endothelial cells and restored by inhibition of proteasomal degradation pathways. The results provide underlying mechanisms for the clinically observed inefficacy of glucocorticoids. The novel combined treatment strategy might help to attenuate trauma-induced brain edema formation and neuronal damage as secondary effects of brain trauma.
Collapse
|
37
|
Shikatani EA, Trifonova A, Mandel ER, Liu STK, Roudier E, Krylova A, Szigiato A, Beaudry J, Riddell MC, Haas TL. Inhibition of proliferation, migration and proteolysis contribute to corticosterone-mediated inhibition of angiogenesis. PLoS One 2012; 7:e46625. [PMID: 23056375 PMCID: PMC3462789 DOI: 10.1371/journal.pone.0046625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 09/07/2012] [Indexed: 12/21/2022] Open
Abstract
The angiostatic nature of pharmacological doses of glucocorticoid steroids is well known. However, the consequences of pathophysiological elevation of endogenous glucocorticoids are not well established. In the current study, we hypothesized that the angiostatic effect of corticosterone, an endogenous glucocorticoid in rodents, occurs through multi-faceted alterations in skeletal muscle microvascular endothelial cell proliferation, migration, and proteolysis. Chronic corticosterone treatment significantly reduced the capillary to fiber ratio in the tibialis anterior muscle compared to that of placebo-treated rats. Corticosterone inhibited endothelial cell sprouting from capillary segments ex vivo. Similarly, 3-dimensional endothelial cell spheroids treated with corticosterone for 48 hours showed evidence of sprout regression and reduced sprout length. Endothelial cell proliferation was reduced in corticosterone treated cells, coinciding with elevated FoxO1 and reduced VEGF production. Corticosterone treated endothelial cells exhibited reduced migration, which correlated with a reduction in RhoA activity. Furthermore, corticosterone treated endothelial cells in both 3-dimensional and monolayer cultures had decreased MMP-2 production and activation resulting in decreased proteolysis by endothelial cells, limiting their angiogenic potential. Promoter assays revealed that corticosterone treatment transcriptionally repressed MMP-2, which may map to a predicted GRE between -1510 and -1386 bp of the MMP-2 promoter. Additionally, Sp1, a known transcriptional activator of MMP-2 was decreased following corticosterone treatment. This study provides new insights into the mechanisms by which pathophysiological levels of endogenous glucocorticoids may exert angiostatic effects.
Collapse
Affiliation(s)
- Eric A. Shikatani
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Anastassia Trifonova
- Department of Biology, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Erin R. Mandel
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Sammy T. K. Liu
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Emilie Roudier
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Anna Krylova
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Andrei Szigiato
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Jacqueline Beaudry
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Michael C. Riddell
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Tara L. Haas
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
- Department of Biology, York University, Toronto, Ontario, Canada
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| |
Collapse
|
38
|
Dietrich J, Rao K, Pastorino S, Kesari S. Corticosteroids in brain cancer patients: benefits and pitfalls. Expert Rev Clin Pharmacol 2012; 4:233-42. [PMID: 21666852 DOI: 10.1586/ecp.11.1] [Citation(s) in RCA: 234] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glucocorticoids have been used for decades in the treatment of brain tumor patients and belong to the most powerful class of agents in reducing tumor-associated edema and minimizing side effects and the risk of encephalopathy in patients undergoing radiation therapy. Unfortunately, corticosteroids are associated with numerous and well-characterized adverse effects, constituting a major challenge in patients requiring long-term application of corticosteroids. Novel antiangiogenic agents, such as bevacizumab (Avastin®), which have been increasingly used in cancer patients, are associated with significant steroid-sparing effects, allowing neuro-oncologists to reduce the overall use of corticosteroids in patients with progressive malignant brain tumors. Recent experimental studies have revealed novel insights into the mechanisms and effects of corticosteroids in cancer patients, including modulation of tumor biology, angiogenesis and steroid-associated neurotoxicity. This article summarizes current concepts of using corticosteroids in brain cancer patients and highlights potential pitfalls in their effects on both tumor and neural progenitor cells.
Collapse
Affiliation(s)
- Jörg Dietrich
- MGH Cancer Center and Center for Regenerative Medicine, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
39
|
Burek M, Salvador E, Förster CY. Generation of an immortalized murine brain microvascular endothelial cell line as an in vitro blood brain barrier model. J Vis Exp 2012:e4022. [PMID: 22951995 PMCID: PMC3486758 DOI: 10.3791/4022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Epithelial and endothelial cells (EC) are building paracellular barriers which protect the tissue from the external and internal environment. The blood-brain barrier (BBB) consisting of EC, astrocyte end-feet, pericytes and the basal membrane is responsible for the protection and homeostasis of the brain parenchyma. In vitro BBB models are common tools to study the structure and function of the BBB at the cellular level. A considerable number of different in vitro BBB models have been established for research in different laboratories to date. Usually, the cells are obtained from bovine, porcine, rat or mouse brain tissue (discussed in detail in the review by Wilhelm et al.). Human tissue samples are available only in a restricted number of laboratories or companies. While primary cell preparations are time consuming and the EC cultures can differ from batch to batch, the establishment of immortalized EC lines is the focus of scientific interest. Here, we present a method for establishing an immortalized brain microvascular EC line from neonatal mouse brain. We describe the procedure step-by-step listing the reagents and solutions used. The method established by our lab allows the isolation of a homogenous immortalized endothelial cell line within four to five weeks. The brain microvascular endothelial cell lines termed cEND (from cerebral cortex) and cerebEND (from cerebellar cortex), were isolated according to this procedure in the Förster laboratory and have been effectively used for explanation of different physiological and pathological processes at the BBB. Using cEND and cerebEND we have demonstrated that these cells respond to glucocorticoid- and estrogen-treatment as well as to pro-infammatory mediators, such as TNFalpha. Moreover, we have studied the pathology of multiple sclerosis and hypoxia on the EC-level. The cEND and cerebEND lines can be considered as a good tool for studying the structure and function of the BBB, cellular responses of ECs to different stimuli or interaction of the EC with lymphocytes or cancer cells.
Collapse
Affiliation(s)
- Malgorzata Burek
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg
| | | | | |
Collapse
|
40
|
|
41
|
Yao LC, Baluk P, Srinivasan RS, Oliver G, McDonald DM. Plasticity of button-like junctions in the endothelium of airway lymphatics in development and inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2561-75. [PMID: 22538088 DOI: 10.1016/j.ajpath.2012.02.019] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 02/03/2012] [Accepted: 02/09/2012] [Indexed: 12/14/2022]
Abstract
Endothelial cells of initial lymphatics have discontinuous button-like junctions (buttons), unlike continuous zipper-like junctions (zippers) of collecting lymphatics and blood vessels. Buttons are thought to act as primary valves for fluid and cell entry into lymphatics. To learn when and how buttons form during development and whether they change in disease, we examined the appearance of buttons in mouse embryos and their plasticity in sustained inflammation. We found that endothelial cells of lymph sacs at embryonic day (E)12.5 and tracheal lymphatics at E16.5 were joined by zippers, not buttons. However, zippers in initial lymphatics decreased rapidly just before birth, as buttons appeared. The proportion of buttons increased from only 6% at E17.5 and 12% at E18.5 to 35% at birth, 50% at postnatal day (P)7, 90% at P28, and 100% at P70. In inflammation, zippers replaced buttons in airway lymphatics at 14 and 28 days after Mycoplasma pulmonis infection of the respiratory tract. The change in lymphatic junctions was reversed by dexamethasone but not by inhibition of vascular endothelial growth factor receptor-3 signaling by antibody mF4-31C1. Dexamethasone also promoted button formation during early postnatal development through a direct effect involving glucocorticoid receptor phosphorylation in lymphatic endothelial cells. These findings demonstrate the plasticity of intercellular junctions in lymphatics during development and inflammation and show that button formation can be promoted by glucocorticoid receptor signaling in lymphatic endothelial cells.
Collapse
Affiliation(s)
- Li-Chin Yao
- Department of Anatomy, Cardiovascular Research Institute, and the Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
42
|
Kielczewski JL, Li Calzi S, Shaw LC, Cai J, Qi X, Ruan Q, Wu L, Liu L, Hu P, Chan-Ling T, Mames RN, Firth S, Baxter RC, Turowski P, Busik JV, Boulton ME, Grant MB. Free insulin-like growth factor binding protein-3 (IGFBP-3) reduces retinal vascular permeability in association with a reduction of acid sphingomyelinase (ASMase). Invest Ophthalmol Vis Sci 2011; 52:8278-86. [PMID: 21931131 DOI: 10.1167/iovs.11-8167] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE To examine the effect of free insulin-like growth factor (IGF) binding protein-3 (IGFBP-3), independent of the effect of insulin-like growth factors, in modulating retinal vascular permeability. METHODS We assessed the ability of a form of IGFBP-3 that does not bind IGF-1 (IGFBP-3NB), to regulate the blood retinal barrier (BRB) using two distinct experimental mouse models, laser-induced retinal vessel injury and vascular endothelial growth factor (VEGF)-induced retinal vascular permeability. Additionally, in vitro studies were conducted. In the animal models, BRB permeability was quantified by intravenous injection of fluorescein labeled serum albumin followed by digital confocal image analysis of retinal flat-mounts. Claudin-5 and vascular endothelial-cadherin (VE-cadherin) localization at interendothelial junctions was studied by immunofluorescence. In vitro changes in transendothelial electrical resistance (TEER) and flux of fluorescent dextran in bovine retinal endothelial monolayers (BREC) were measured after IGFBP-3NB treatment. Acid (ASMase) and neutral (NSMase) sphingomyelinase mRNA levels and activity were measured in mouse retinas. RESULTS Four days postinjury, laser-injured mouse retinas injected with IGFBP-3NB plasmid demonstrated reduced vascular permeability compared with retinas of laser-injured mouse retinas injected with control plasmid. IGFBP-3NB administration resulted in a significant decrease in laser injury-associated increases in ASMase and NSMase mRNA and activity when compared with laser alone treated mice. In vivo, intravitreal injection of IGFBP-3NB reduced vascular leakage associated with intravitreal VEGF injection. IGFBP-3NB partially restored VEGF-induced in vivo permeability and dissociation of claudin-5 and VE-cadherin at junctional complexes. When IGFBP-3NB was applied basally to bovine retinal endothelial cells (BREC) in vitro, TEER increased and macromolecular flux decreased. CONCLUSIONS Intravitreal administration of IGFBP-3NB preserves junctional integrity in the presence of VEGF or laser injury by reducing BRB permeability in part by modulating sphingomyelinase levels.
Collapse
Affiliation(s)
- Jennifer L Kielczewski
- Departments of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610-0267, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Gómez‐González B, Larios HM, Escobar A. Increased transvascular transport of WGA‐peroxidase after chronic perinatal stress in the hippocampal microvasculature of the rat. Int J Dev Neurosci 2011; 29:839-46. [DOI: 10.1016/j.ijdevneu.2011.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 07/08/2011] [Accepted: 08/08/2011] [Indexed: 12/20/2022] Open
Affiliation(s)
- Beatriz Gómez‐González
- Dept. Cell Biology and PhysiologyInstituto de Investigaciones BiomédicasUniversidad Nacional Autónoma de MéxicoCiudad Universitaria04510Mexico CityMexico
| | - Horacio Merchant Larios
- Dept. Cell Biology and PhysiologyInstituto de Investigaciones BiomédicasUniversidad Nacional Autónoma de MéxicoCiudad Universitaria04510Mexico CityMexico
| | - Alfonso Escobar
- Dept. Cell Biology and PhysiologyInstituto de Investigaciones BiomédicasUniversidad Nacional Autónoma de MéxicoCiudad Universitaria04510Mexico CityMexico
| |
Collapse
|
44
|
Neuwelt EA, Bauer B, Fahlke C, Fricker G, Iadecola C, Janigro D, Leybaert L, Molnar Z, O’Donnell M, Povlishock J, Saunders N, Sharp F, Stanimirovic D, Watts R, Drewes L. Engaging neuroscience to advance translational research in brain barrier biology. Nat Rev Neurosci 2011; 12:169-82. [PMID: 21331083 PMCID: PMC3335275 DOI: 10.1038/nrn2995] [Citation(s) in RCA: 392] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The delivery of many potentially therapeutic and diagnostic compounds to specific areas of the brain is restricted by brain barriers, of which the most well known are the blood-brain barrier (BBB) and the blood-cerebrospinal fluid (CSF) barrier. Recent studies have shown numerous additional roles of these barriers, including an involvement in neurodevelopment, in the control of cerebral blood flow, and--when barrier integrity is impaired--in the pathology of many common CNS disorders such as Alzheimer's disease, Parkinson's disease and stroke.
Collapse
Affiliation(s)
- Edward A. Neuwelt
- Oregon Health & Science University, Portland, Oregon
- Portland Veterans Affairs Medical Center, Portland, Oregon
| | | | | | | | | | | | | | | | | | | | | | - Frank Sharp
- University of California at Davis, Davis, California
| | | | - Ryan Watts
- Genentech, Inc., South San Francisco, California
| | | |
Collapse
|
45
|
Kimura K, Teranishi S, Kawamoto K, Nishida T. Protective effect of dexamethasone against hypoxia-induced disruption of barrier function in human corneal epithelial cells. Exp Eye Res 2011; 92:388-93. [PMID: 21354133 DOI: 10.1016/j.exer.2011.02.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 01/17/2011] [Accepted: 02/17/2011] [Indexed: 01/01/2023]
Abstract
The corneal epithelium functions as a barrier to protect the cornea from external agents such as infectious organisms and toxins and thereby contributes to corneal homeostasis. The barrier function of epithelia is dependent on the formation of tight and adherens junctions between adjacent epithelial cells. We have previously shown that hypoxia disrupts the barrier function of cultured human corneal epithelial (HCE) cells by affecting tight junctions. We have now examined the effect of dexamethasone on this barrier disruption induced by hypoxia in HCE cells. Measurement of transepithelial electrical resistance revealed that the hypoxia-induced decrease in the barrier function of HCE cells was inhibited by dexamethasone in a concentration-dependent manner. The hypoxia-induced loss of the tight junction protein ZO-1 from the borders of adjacent HCE cells (as revealed by immunofluorescence analysis) as well as the hypoxia-induced down-regulation of ZO-1 expression (as revealed by immunoblot analysis) were also inhibited by dexamethasone, whereas this drug had no effect on the expression or distribution of the tight junction protein occludin or of the adherens junction proteins E-cadherin and β-catenin. Moreover, dexamethasone attenuated the reorganization of the actin cytoskeleton, the formation of focal adhesions, and the up-regulation of myosin light chain kinase expression induced by hypoxia in HCE cells. Our results thus suggest that dexamethasone protects corneal epithelial cells from the hypoxia-induced disruption of barrier function by maintaining the distribution and expression of ZO-1 as well as the organization of the actin cytoskeleton.
Collapse
Affiliation(s)
- Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan.
| | | | | | | |
Collapse
|
46
|
Shen W, Li S, Chung SH, Zhu L, Stayt J, Su T, Couraud PO, Romero IA, Weksler B, Gillies MC. Tyrosine phosphorylation of VE-cadherin and claudin-5 is associated with TGF-β1-induced permeability of centrally derived vascular endothelium. Eur J Cell Biol 2010; 90:323-32. [PMID: 21168935 DOI: 10.1016/j.ejcb.2010.10.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 10/21/2010] [Accepted: 10/21/2010] [Indexed: 01/25/2023] Open
Abstract
Breakdown of the inner blood-retinal barrier and the blood-brain barrier is associated with changes in tight and adherens junction-associated proteins that link vascular endothelial cells. This study aimed to test the hypothesis that transforming growth factor (TGF)-β1 increases the paracellular permeability of vascular endothelial monolayers through tyrosine phosphorylation of VE-cadherin and claudin-5. Bovine retinal and human brain capillary endothelial cells were grown as monolayers on coated polycarbonate membranes. Paracellular permeability was studied by measuring the equilibration of (14)C-inulin or fluorescence-labelled dextran. Changes in VE-cadherin and claudin-5 expression were studied by immunocytochemistry (ICC) and quantified by cell-based enzyme linked immunosorbent assays (ELISA). Tyrosine phosphorylation of VE-cadherin and claudin-5 was studied by ICC, immunoprecipitation and Western blotting. We found that exposure of endothelial cells to TGF-β1 caused a dose-dependent increase in paracellular permeability as reflected by increases in the equilibration of (14)C-inulin. This effect was enhanced by the tyrosine phosphatase inhibitor orthovanadate and attenuated by the tyrosine kinase inhibitor lavendustin A. ICC and cell-based ELISA revealed that TGF-β1 induced both dose- and time-dependent decreases in VE-cadherin and claudin-5 expression. Assessment of cell viability indicated that changes in these junction-associated proteins were not due to endothelial death or injury. ICC revealed that tyrosine phosphorylation of endothelial monolayers was greatly enhanced by TGF-β1 treatment, and immunoprecipitation of cell lysates showed increased tyrosine phosphorylation of VE-cadherin and claudin-5. Our results suggest that tyrosine phosphorylation of VE-cadherin and claudin-5 is involved in the increased paracellular permeability of central nervous system-derived vascular endothelium induced by TGF-β1.
Collapse
Affiliation(s)
- Weiyong Shen
- Save Sight Institute, the University of Sydney, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yang L, Zhang H, Fawcett JP, Mikov M, Tucker IG. Effect of bile salts on the transport of morphine-6-glucuronide in rat brain endothelial cells. J Pharm Sci 2010; 100:1516-24. [PMID: 24081474 DOI: 10.1002/jps.22390] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 09/06/2010] [Accepted: 09/30/2010] [Indexed: 11/09/2022]
Abstract
Bile salts are known to enhance the permeability of biological barriers but little is known about their effects on drug permeability across the blood-brain barrier (BBB). In this paper, the rat brain endothelial 4 (RBE4) cell monolayer incubated with astrocyte-conditioned medium was used as an in vitro model of the BBB to investigate the effects of cholate (C), 12-monoketocholate (MKC), deoxycholate (DC), and taurocholate (TC) on the transport of the hydrophilic drug, morphine-6-glucuronide (M6G). C, MKC, and TC at a concentration of 5 mM each and DC at 1 mM increased the permeability of M6G through the paracellular pathway based on a similar permeability pattern to that of sucrose. RBE4 cell uptake of M6G was unaffected by 5 mM C and TC, whereas 1 mM DC dramatically increased it due to an effect shown to be cytotoxicity as measured by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Surprisingly, 1 mM MKC significantly increased M6G uptake without any cytotoxicity. In summary, all bile salts increased paracellular permeation of M6G but MKC also enhanced transcellular transport with little cytotoxicity. MKC appears to have the potential to modulate biophysical properties of the cell membrane or membrane-bound transporters and may therefore enhance drug delivery to the brain.
Collapse
Affiliation(s)
- Lin Yang
- School of Pharmacy, University of Otago, Dunedin, New Zealand.
| | | | | | | | | |
Collapse
|
48
|
Blecharz KG, Haghikia A, Stasiolek M, Kruse N, Drenckhahn D, Gold R, Roewer N, Chan A, Förster CY. Glucocorticoid effects on endothelial barrier function in the murine brain endothelial cell line cEND incubated with sera from patients with multiple sclerosis. Mult Scler 2010; 16:293-302. [DOI: 10.1177/1352458509358189] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Compromised blood—brain barrier integrity is a major hallmark of active multiple sclerosis (MS). Alterations in brain endothelial tight junction protein and gene expression occur early during neuroinflammation but there is little known about the underlying mechanisms. In this study, we analysed barrier compromising effects of sera from MS patients and barrier restoring effects of glucocorticoids on blood—brain barrier integrity in vitro. cEND murine brain microvascular endothelial cell monolayers were incubated with sera from patients in active phase of disease or in relapse. Data were compared with effects of the glucocorticoid dexamethasone alone or in combination with MS sera on barrier integrity. Tight junction protein levels and gene expression were evaluated concomitant with barrier integrity. We reveal downregulation of claudin-5 and occludin protein and mRNA and an accompanying upregulation in expression of matrix metalloproteinase MMP-9 after incubation with serum from active disease and remission and also a minor reconstitution of barrier functions related to dexamethasone treatment. Moreover, we for the first time describe downregulation of claudin-5 and occludin protein after incubation of cEND cells with sera from patients in remission phase of MS. Our findings reveal direct and differential effects of MS sera on blood-brain barrier integrity.
Collapse
Affiliation(s)
- Kinga G Blecharz
- Department of Anaesthesiology and Critical Care, University of Würzburg, Würzburg, Germany
| | - Aiden Haghikia
- Department of Neurology, Ruhr University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Mariusz Stasiolek
- Department of Neurology, Ruhr University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Niels Kruse
- Institute of Multiple Sclerosis Research, University of Göttingen and Gemeinnützige Hertie-Stiftung, Göttingen, Germany
| | - Detlev Drenckhahn
- Institute of Anatomy and Cell Biology , University of Würzburg, Würzburg, Germany
| | - Ralf Gold
- Department of Neurology, Ruhr University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Norbert Roewer
- Department of Anaesthesiology and Critical Care, University of Würzburg, Würzburg, Germany
| | - Andrew Chan
- Department of Neurology, Ruhr University Bochum, St. Josef-Hospital, Bochum, Germany
| | - Carola Y Förster
- Department of Anaesthesiology and Critical Care, University of Würzburg, Würzburg, Germany,
| |
Collapse
|
49
|
Altered functional development of the blood–brain barrier after early life stress in the rat. Brain Res Bull 2009; 79:376-87. [DOI: 10.1016/j.brainresbull.2009.05.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 05/09/2009] [Accepted: 05/13/2009] [Indexed: 11/22/2022]
|
50
|
Eyal S, Hsiao P, Unadkat JD. Drug interactions at the blood-brain barrier: fact or fantasy? Pharmacol Ther 2009; 123:80-104. [PMID: 19393264 DOI: 10.1016/j.pharmthera.2009.03.017] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Accepted: 03/20/2009] [Indexed: 12/24/2022]
Abstract
There is considerable interest in the therapeutic and adverse outcomes of drug interactions at the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB). These include altered efficacy of drugs used in the treatment of CNS disorders, such as AIDS dementia and malignant tumors, and enhanced neurotoxicity of drugs that normally penetrate poorly into the brain. BBB- and BCSFB-mediated interactions are possible because these interfaces are not only passive anatomical barriers, but are also dynamic in that they express a variety of influx and efflux transporters and drug metabolizing enzymes. Based on studies in rodents, it has been widely postulated that efflux transporters play an important role at the human BBB in terms of drug delivery. Furthermore, it is assumed that chemical inhibition of transporters or their genetic ablation in rodents is predictive of the magnitude of interaction to be expected at the human BBB. However, studies in humans challenge this well-established paradigm and claim that such drug interactions will be lesser in magnitude but yet may be clinically significant. This review focuses on current known mechanisms of drug interactions at the blood-brain and blood-CSF barriers and the potential impact of such interactions in humans. We also explore whether such drug interactions can be predicted from preclinical studies. Defining the mechanisms and the impact of drug-drug interactions at the BBB is important for improving efficacy of drugs used in the treatment of CNS disorders while minimizing their toxicity as well as minimizing neurotoxicity of non-CNS drugs.
Collapse
Affiliation(s)
- Sara Eyal
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195, USA
| | | | | |
Collapse
|