1
|
Zhan JH, Wei J, Liu YJ, Wang PX, Zhu XY. Sepsis-associated endothelial glycocalyx damage: a review of animal models, clinical evidence, and molecular mechanisms. Int J Biol Macromol 2025; 295:139548. [PMID: 39788232 DOI: 10.1016/j.ijbiomac.2025.139548] [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: 07/03/2024] [Revised: 12/21/2024] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
In the mammalian cardiovascular system, endothelial glycocalyx is a gel-like layer that covers the luminal surface of endothelial cells (ECs) and plays crucial roles in vascular homeostasis, permeability and leukocyte adhesion. Degradation of this structure occurs early in sepsis and becomes accordingly dysfunctional. In severe cases, it is not self-regulated by the organism. However, the relationship between the glycocalyx and the occurrence and development of sepsis remains poorly understood. One possibility is that thinned glycocalyx promotes leukocyte recognition and adhesion, thereby facilitating the elimination of pathogens from infected areas. This may represent a protective mechanism developed by the organism during through evolutionary processes. However, if the damage persists and disrupts the dynamic balance of the microcirculation, interstitial edema or organ failure can occur. Thus, we asked the questions, what is the precise composition and structure of the glycocalyx? How is it degraded? What animal models are available to study the relationship between the glycocalyx and sepsis? What glycocalyx biomarkers are found in the blood of patients with sepsis? To determine whether sepsis can be treated by interfering with the glycocalyx, this study provides a systematic summary and discussion of the latest progress in addressing these questions.
Collapse
Affiliation(s)
- Jun-Hui Zhan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China; Department of Physiology, Naval Medical University, Shanghai 200433, China
| | - Juan Wei
- School of Sports and Health, Nanjing Sport Institute, Nanjing 210014, China
| | - Yu-Jian Liu
- School of Kinesiology, The Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China
| | - Peng-Xiang Wang
- Department of Physiology, Naval Medical University, Shanghai 200433, China.
| | - Xiao-Yan Zhu
- Department of Physiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
2
|
Jhaveri JR, Khare P, Paul Pinky P, Kamte YS, Chandwani MN, Milosevic J, Abraham N, Sun M, Stolz DB, Dave KM, Zheng SY, O'Donnell L, Manickam DS. Low pinocytic brain endothelial cells primarily utilize membrane fusion to internalize extracellular vesicles. Eur J Pharm Biopharm 2024; 204:114500. [PMID: 39303949 DOI: 10.1016/j.ejpb.2024.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Extracellular vesicles (EVs) are an emerging class of drug carriers and are primarily reported to be internalized into recipient cells via a combination of endocytic routes such as clathrin-mediated, caveolae-mediated and macropinocytosis pathways. In this work, (1) we investigated potential effects of homotypic vs. heterotypic interactions by studying the cellular uptake of homologous EVs (EV donor cells and recipient cells of the same type) vs. heterologous EVs (EV donor cells and recipient cells of different types) and (2) determined the route of EV internalization into low pinocytic/hard-to-deliver cell models such as brain endothelial cells (BECs). Homotypic interactions led to a greater extent of uptake into the recipient BECs compared to heterotypic interactions. However, we did not see a complete reduction in EV uptake into recipient BECs when endocytic pathways were blocked using pharmacological inhibitors and our findings from a R18-based fusion assay suggest that EVs primarily use membrane fusion to enter low-pinocytic recipient BECs instead of relying on endocytosis. Lipophilic PKH67 dye-labeled EVs but not intravesicular esterase-activated calcein ester-labeled EVs severely reduced particle uptake into BECs while phagocytic macrophages internalized EVs labeled with both dyes to comparable extents. Our results also highlight the importance of carefully choosing labeling dye chemistry to study EV uptake, especially in the case of low pinocytic cells such as BECs.
Collapse
Affiliation(s)
- Jhanvi R Jhaveri
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Purva Khare
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Paromita Paul Pinky
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Yashika S Kamte
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Manisha N Chandwani
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Jadranka Milosevic
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States; Captis Diagnostics Inc., Pittsburgh, PA, United States
| | - Nevil Abraham
- Unified Flow Core, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ming Sun
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical School, Pittsburgh, PA, United States
| | - Kandarp M Dave
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Si-Yang Zheng
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Lauren O'Donnell
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
| | - Devika S Manickam
- Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States.
| |
Collapse
|
3
|
Balistreri CR, Di Giorgi L, Monastero R. Focus of endothelial glycocalyx dysfunction in ischemic stroke and Alzheimer's disease: Possible intervention strategies. Ageing Res Rev 2024; 99:102362. [PMID: 38830545 DOI: 10.1016/j.arr.2024.102362] [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: 04/22/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
The integrity of the endothelial glycocalyx (eGCX), a mixture of carbohydrates attached to proteins expressed on the surface of blood vessel endothelial cells (EC), is critical for the maintenance of homeostasis of the cardiovascular system and all systems of the human body, the endothelium being the critical component of the stroma of all tissues. Consequently, dysfunction of eGCX results in a dysfunctional cardiovascular wall and severe downstream cardiovascular events, which contribute to the onset of cardio- and cerebrovascular diseases and neurodegenerative disorders, as well as other age-related diseases (ARDs). The key role of eGCX dysfunction in the onset of ARDs is examined here, with a focus on the most prevalent neurological diseases: ischemic stroke and Alzheimer's disease. Furthermore, the advantages and limitations of some treatment strategies for anti-eGCX dysfunction are described, ranging from experimental drug therapies, which need to be better tested and explored not only in animal models but also in humans, as well as reprogramming, the use of nutraceuticals, which are emerging as regenerative and new approaches. The promotion of these strategies is essential to keep eGCX and endothelium healthy, as is the development of intravital (e.g., intravascular) tools to estimate eGCX health status and treatment efficacy, which could lead to advanced solutions to address ARDs.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Cellular, Molecular and Clinical Pathological Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo 90134, Italy.
| | - Lucia Di Giorgi
- Memory and Parkinson's disease Center Policlinico "Paolo Giaccone", Palermo, and Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Via La Loggia 1, Palermo 90129, Italy
| | - Roberto Monastero
- Memory and Parkinson's disease Center Policlinico "Paolo Giaccone", Palermo, and Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Via La Loggia 1, Palermo 90129, Italy.
| |
Collapse
|
4
|
O’Hare N, Millican K, Ebong EE. Unraveling neurovascular mysteries: the role of endothelial glycocalyx dysfunction in Alzheimer's disease pathogenesis. Front Physiol 2024; 15:1394725. [PMID: 39027900 PMCID: PMC11254711 DOI: 10.3389/fphys.2024.1394725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/27/2024] [Indexed: 07/20/2024] Open
Abstract
While cardiovascular disease, cancer, and human immunodeficiency virus (HIV) mortality rates have decreased over the past 20 years, Alzheimer's Disease (AD) deaths have risen by 145% since 2010. Despite significant research efforts, effective AD treatments remain elusive due to a poorly defined etiology and difficulty in targeting events that occur too downstream of disease onset. In hopes of elucidating alternative treatment pathways, now, AD is commonly being more broadly defined not only as a neurological disorder but also as a progression of a variety of cerebrovascular pathologies highlighted by the breakdown of the blood-brain barrier. The endothelial glycocalyx (GCX), which is an essential regulator of vascular physiology, plays a crucial role in the function of the neurovascular system, acting as an essential vascular mechanotransducer to facilitate ultimate blood-brain homeostasis. Shedding of the cerebrovascular GCX could be an early indication of neurovascular dysfunction and may subsequently progress neurodegenerative diseases like AD. Recent advances in in vitro modeling, gene/protein silencing, and imaging techniques offer new avenues of scrutinizing the GCX's effects on AD-related neurovascular pathology. Initial studies indicate GCX degradation in AD and other neurodegenerative diseases and have begun to demonstrate a possible link to GCX loss and cerebrovascular dysfunction. This review will scrutinize the GCX's contribution to known vascular etiologies of AD and propose future work aimed at continuing to uncover the relationship between GCX dysfunction and eventual AD-associated neurological deterioration.
Collapse
Affiliation(s)
- Nicholas O’Hare
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
| | - Karina Millican
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Eno E. Ebong
- Department of Chemical Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, Northeastern University, Boston, MA, United States
- Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
5
|
Nguyen TK, Paone S, Baxter AA, Mayfosh AJ, Phan TK, Chan E, Peter K, Poon IKH, Thomas SR, Hulett MD. Heparanase promotes the onset and progression of atherosclerosis in apolipoprotein E gene knockout mice. Atherosclerosis 2024; 392:117519. [PMID: 38581737 DOI: 10.1016/j.atherosclerosis.2024.117519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/31/2024] [Accepted: 03/12/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis is the primary underlying cause of myocardial infarction and stroke, which are the major causes of death globally. Heparanase (Hpse) is a pro-inflammatory extracellular matrix degrading enzyme that has been implicated in atherogenesis. However, to date the precise roles of Hpse in atherosclerosis and its mechanisms of action are not well defined. This study aims to provide new insights into the contribution of Hpse in different stages of atherosclerosis in vivo. METHODS We generated Hpse gene-deficient mice on the atherosclerosis-prone apolipoprotein E gene knockout (ApoE-/-) background to investigate the impact of Hpse gene deficiency on the initiation and progression of atherosclerosis after 6 and 14 weeks high-fat diet feeding, respectively. Atherosclerotic lesion development, blood serum profiles, lesion composition and aortic immune cell populations were evaluated. RESULTS Hpse-deficient mice exhibited significantly reduced atherosclerotic lesion burden in the aortic sinus and aorta at both time-points, independent of changes in plasma cholesterol levels. A significant reduction in the necrotic core size and an increase in smooth muscle cell content were also observed in advanced atherosclerotic plaques of Hpse-deficient mice. Additionally, Hpse deficiency reduced circulating and aortic levels of VCAM-1 at the initiation and progression stages of disease and circulating MCP-1 levels in the initiation but not progression stage. Moreover, the aortic levels of total leukocytes and dendritic cells in Hpse-deficient ApoE-/- mice were significantly decreased compared to control ApoE-/-mice at both disease stages. CONCLUSIONS This study identifies Hpse as a key pro-inflammatory enzyme driving the initiation and progression of atherosclerosis and highlighting the potential of Hpse inhibitors as novel anti-inflammatory treatments for cardiovascular disease.
Collapse
Affiliation(s)
- Tien K Nguyen
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Stephanie Paone
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Amy A Baxter
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Alyce J Mayfosh
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Thanh Kha Phan
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Enoch Chan
- Department of Pathology, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, 3004, Australia
| | - Ivan K H Poon
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Shane R Thomas
- Department of Pathology, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Mark D Hulett
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia.
| |
Collapse
|
6
|
Song YX, Li X, Nie SD, Hu ZX, Zhou D, Sun DY, Zhou GY, Wang Y, Liu JJ, Song T, Wang S. Extracellular vesicles released by glioma cells are decorated by Annexin A2 allowing for cellular uptake via heparan sulfate. Cancer Gene Ther 2023; 30:1156-1166. [PMID: 37231059 DOI: 10.1038/s41417-023-00627-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/20/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023]
Abstract
Extracellular vesicles (EVs) play a crucial role in regulating cell behavior by delivering their cargo to target cells. However, the mechanisms underlying EV-cell interactions are not well understood. Previous studies have shown that heparan sulfate (HS) on target cell surfaces can act as receptors for exosomes uptake, but the ligand for HS on EVs has not been identified. In this study, we isolated EVs from glioma cell lines and glioma patients and identified Annexin A2 (AnxA2) on EVs as a key HS-binding ligand and mediator of EV-cell interactions. Our findings suggest that HS plays a dual role in EV-cell interactions, where HS on EVs captures AnxA2, and on target cells, it acts as a receptor for AnxA2. Removal of HS from the EV surface inhibits EV-target cell interaction by releasing AnxA2. Furthermore, we found that AnxA2-mediated binding of EVs to vascular endothelial cells promotes angiogenesis, and that antibody against AnxA2 inhibited the ability of glioma-derived EVs to stimulate angiogenesis by reducing the uptake of EVs. Our study also suggests that the AnxA2-HS interaction may accelerate the glioma-derived EVs-mediated angiogenesis and that combining AnxA2 on glioma cells with HS on endothelial cells may effectively improve the prognosis evaluation of glioma patients.
Collapse
Affiliation(s)
- Yu-Xi Song
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, Changsha, China
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Xin Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China.
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, Changsha, China.
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Sheng-Dan Nie
- Institute of Clinical Medicine, Hunan provincial people's hospital, the first affiliated hospital of Hunan Normal University, Changsha, China
| | - Zhong-Xu Hu
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Di Zhou
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Ding-Ya Sun
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Gao-Ya Zhou
- Department of Neurology, Brain hospital of Hunan Province, Changsha, China
| | - Ying Wang
- Department of Pathology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jia-Jia Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Tao Song
- Department of Neurosurgery, Xiang-Ya Hospital, Central South University, Changsha, China.
| | - Shan Wang
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| |
Collapse
|
7
|
Lindsay SL, McCanney GA, Zhan J, Scheld M, Smith RS, Goodyear CS, Yates EA, Kipp M, Turnbull JE, Barnett SC. Low sulfated heparan sulfate mimetic differentially affects repair in immune-mediated and toxin-induced experimental models of demyelination. Glia 2023; 71:1683-1698. [PMID: 36945189 PMCID: PMC10952530 DOI: 10.1002/glia.24363] [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: 11/03/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
There is an urgent need for therapies that target the multicellular pathology of central nervous system (CNS) disease. Modified, nonanticoagulant heparins mimic the heparan sulfate glycan family and are known regulators of multiple cellular processes. In vitro studies have demonstrated that low sulfated modified heparin mimetics (LS-mHeps) drive repair after CNS demyelination. Herein, we test LS-mHep7 (an in vitro lead compound) in experimental autoimmune encephalomyelitis (EAE) and cuprizone-induced demyelination. In EAE, LS-mHep7 treatment resulted in faster recovery and rapidly reduced inflammation which was accompanied by restoration of animal weight. LS-mHep7 treatment had no effect on remyelination or on OLIG2 positive oligodendrocyte numbers within the corpus callosum in the cuprizone model. Further in vitro investigation confirmed that LS-mHep7 likely mediates its pro-repair effect in the EAE model by sequestering inflammatory cytokines, such as CCL5 which are upregulated during immune-mediated inflammatory attacks. These data support the future clinical translation of this next generation modified heparin as a treatment for CNS diseases with active immune system involvement.
Collapse
Affiliation(s)
- Susan L. Lindsay
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - George A. McCanney
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - Jiangshan Zhan
- Institute of AnatomyUniversity of RostockGertrudenstrasse 918057RostockGermany
| | - Miriam Scheld
- Institute of Neuroanatomy, Faculty of MedicineRWTH Aachen University52074AachenGermany
| | - Rebecca Sherrard Smith
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - Carl S. Goodyear
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - Edwin A. Yates
- Institute of Systems, Molecules and Integrative BiologyUniversity of LiverpoolLiverpoolL69 7ZBUK
| | - Markus Kipp
- Institute of AnatomyUniversity of RostockGertrudenstrasse 918057RostockGermany
| | - Jeremy E. Turnbull
- Institute of Systems, Molecules and Integrative BiologyUniversity of LiverpoolLiverpoolL69 7ZBUK
- Centre for GlycosciencesKeele UniversityKeeleST5 5BGUK
| | - Susan C. Barnett
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| |
Collapse
|
8
|
Hudák A, Letoha A, Vizler C, Letoha T. Syndecan-3 as a Novel Biomarker in Alzheimer's Disease. Int J Mol Sci 2022; 23:3407. [PMID: 35328830 PMCID: PMC8955174 DOI: 10.3390/ijms23063407] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/17/2022] Open
Abstract
Early diagnosis of Alzheimer's disease (AD) is of paramount importance in preserving the patient's mental and physical health in a fairly manageable condition for a longer period. Reliable AD detection requires novel biomarkers indicating central nervous system (CNS) degeneration in the periphery. Members of the syndecan family of transmembrane proteoglycans are emerging new targets in inflammatory and neurodegenerative disorders. Reviewing the growing scientific evidence on the involvement of syndecans in the pathomechanism of AD, we analyzed the expression of the neuronal syndecan, syndecan-3 (SDC3), in experimental models of neurodegeneration. Initial in vitro studies showed that prolonged treatment of tumor necrosis factor-alpha (TNF-α) increases SDC3 expression in model neuronal and brain microvascular endothelial cell lines. In vivo studies revealed elevated concentrations of TNF-α in the blood and brain of APPSWE-Tau transgenic mice, along with increased SDC3 concentration in the brain and the liver. Primary brain endothelial cells and peripheral blood monocytes isolated from APPSWE-Tau mice exhibited increased SDC3 expression than wild-type controls. SDC3 expression of blood-derived monocytes showed a positive correlation with amyloid plaque load in the brain, demonstrating that SDC3 on monocytes is a good indicator of amyloid pathology in the brain. Given the well-established role of blood tests, the SDC3 expression of monocytes could serve as a novel biomarker for early AD detection.
Collapse
Affiliation(s)
| | - Annamária Letoha
- Albert Szent-Györgyi Clinical Center, Department of Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary;
| | - Csaba Vizler
- Biological Research Centre, Institute of Biochemistry, H-6726 Szeged, Hungary;
| | | |
Collapse
|
9
|
Kim HJ, Kim HS, Hong YH. Sulfatase 1 and sulfatase 2 as novel regulators of macrophage antigen presentation and phagocytosis. Yeungnam Univ J Med 2021; 38:326-336. [PMID: 34157797 PMCID: PMC8688788 DOI: 10.12701/yujm.2021.01025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022] Open
Abstract
Background Sulfation of heparan sulfate proteoglycans (HSPGs) is critical for the binding and signaling of ligands that mediate inflammation. Extracellular 6-O-endosulfatases regulate posttranslational sulfation levels and patterns of HSPGs. In this study, extracellular 6-O-endosulfatases, sulfatase (Sulf)-1 and Sulf-2, were evaluated for their expression and function in inflammatory cells and tissues. Methods Harvested human peripheral blood mononuclear cells were treated with phytohemagglutinin and lipopolysaccharide, and murine peritoneal macrophages were stimulated with interleukin (IL)-1β for the evaluation of Sulf-1 and Sulf-2 expression. Sulf expression in inflammatory cells was examined in the human rheumatoid arthritis (RA) synovium by immunofluorescence staining. The antigen presentation and phagocytic activities of macrophages were compared according to the expression state of Sulfs. Sulfs-knockdown macrophages and Sulfs-overexpressing macrophages were generated using small interfering RNAs and pcDNA3.1 plasmids for Sulf-1 and Sulf-2, respectively. Results Lymphocytes and monocytes showed weak Sulf expression, which remained unaffected by IL-1β. However, peritoneal macrophages showed increased expression of Sulfs upon stimulation with IL-1β. In human RA synovium, two-colored double immunofluorescent staining of Sulfs and CD68 revealed active upregulation of Sulfs in macrophages of inflamed tissues, but not in lymphocytes of lymphoid follicles. Macrophages are professional antigen-presenting cells. The antigen presentation and phagocytic activities of macrophages were dependent on the level of Sulf expression, suppressed in Sulfs-knockdown macrophages, and enhanced in Sulfs-overexpressing macrophages. Conclusion The results demonstrate that upregulation of Sulfs in macrophages occurs in response to inflammation, and Sulfs actively regulate the antigen presentation and phagocytic activities of macrophages as novel immune regulators.
Collapse
Affiliation(s)
- Hyun-Je Kim
- Division of Rheumatology, Department of Internal Medicine, CHA University, CHA Gumi Medical Center, Gumi, Korea
| | - Hee-Sun Kim
- Department of Microbiology, Yeungnam University College of Medicine, Daegu, Korea
| | - Young-Hoon Hong
- Division of Rheumatology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| |
Collapse
|
10
|
Ivan DC, Walthert S, Berve K, Steudler J, Locatelli G. Dwellers and Trespassers: Mononuclear Phagocytes at the Borders of the Central Nervous System. Front Immunol 2021; 11:609921. [PMID: 33746939 PMCID: PMC7973121 DOI: 10.3389/fimmu.2020.609921] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/29/2020] [Indexed: 01/02/2023] Open
Abstract
The central nervous system (CNS) parenchyma is enclosed and protected by a multilayered system of cellular and acellular barriers, functionally separating glia and neurons from peripheral circulation and blood-borne immune cells. Populating these borders as dynamic observers, CNS-resident macrophages contribute to organ homeostasis. Upon autoimmune, traumatic or neurodegenerative inflammation, these phagocytes start playing additional roles as immune regulators contributing to disease evolution. At the same time, pathological CNS conditions drive the migration and recruitment of blood-borne monocyte-derived cells across distinct local gateways. This invasion process drastically increases border complexity and can lead to parenchymal infiltration of blood-borne phagocytes playing a direct role both in damage and in tissue repair. While recent studies and technical advancements have highlighted the extreme heterogeneity of these resident and CNS-invading cells, both the compartment-specific mechanism of invasion and the functional specification of intruding and resident cells remain unclear. This review illustrates the complexity of mononuclear phagocytes at CNS interfaces, indicating how further studies of CNS border dynamics are crucially needed to shed light on local and systemic regulation of CNS functions and dysfunctions.
Collapse
|
11
|
Ito S, Ozaki T, Morozumi M, Imagama S, Kadomatsu K, Sakamoto K. Enoxaparin promotes functional recovery after spinal cord injury by antagonizing PTPRσ. Exp Neurol 2021; 340:113679. [PMID: 33662380 DOI: 10.1016/j.expneurol.2021.113679] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 02/03/2023]
Abstract
The receptor-type protein tyrosine phosphatase sigma (PTPRσ) regulates axonal regeneration/sprouting as a molecular switch in response to glycan ligands. Cell surface heparan sulfate oligomerizes PTPRσ and inactivates its enzymatic activity, which in turn promotes axonal growth. In contrast, matrix-associated chondroitin sulfate monomerizes PTPRσ and activates it. This leads to dephosphorylation of its specific substrates, such as cortactin, resulting in a failure of axonal regeneration after injury. However, this molecular switch model has never been challenged in a clinical situation. In this study, we demonstrated that enoxaparin, a globally approved anticoagulant consisting of heparin oligosaccharides with an average molecular weight of 45 kDa, induced clustering and inactivated PTPRσ in vitro. Enoxaparin induced PTPRσ clustering, and counteracted PTPRσ-mediated dephosphorylation of cortactin, which was shown to be important for inhibition of axonal regeneration. Systemic administration of enoxaparin promoted anatomical recovery after both optic nerve and spinal cord injuries in rats at clinically tolerated doses. Moreover, enoxaparin promoted recovery of motor function without obvious hemorrhage. Collectively, our data provide a new strategy for the treatment of traumatic axonal injury.
Collapse
Affiliation(s)
- Sadayuki Ito
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Department of Orthopedics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomoya Ozaki
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Masayoshi Morozumi
- Departments of Orthopedics, Kariya Toyota General Hospital, 15-5 Sumiyoshi-cho, Kariya, 448-8505, Japan
| | - Shiro Imagama
- Department of Orthopedics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan.
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan.
| |
Collapse
|
12
|
Joshi BS, Zuhorn IS. Heparan sulfate proteoglycan-mediated dynamin-dependent transport of neural stem cell exosomes in an in vitro blood-brain barrier model. Eur J Neurosci 2020; 53:706-719. [PMID: 32939863 PMCID: PMC7891616 DOI: 10.1111/ejn.14974] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/10/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022]
Abstract
Drug delivery to the brain is greatly hampered by the presence of the blood–brain barrier (BBB) which tightly regulates the passage of molecules from blood to brain and vice versa. Nanocarriers, in which drugs can be encapsulated, can move across the blood–brain barrier (BBB) via the process of transcytosis, thus showing promise to improve drug delivery to the brain. Here, we demonstrate the use of natural nanovesicles, that is, exosomes, derived from C17.2 neural stem cells (NSCs) to efficiently carry a protein cargo across an in vitro BBB model consisting of human brain microvascular endothelial cells. We show that the exosomes are primarily taken up in brain endothelial cells via endocytosis, while heparan sulfate proteoglycans (HSPGs) act as receptors. Taken together, our data support the view that NSC exosomes may act as biological nanocarriers for efficient passage across the BBB. Nanomedicines that target HSPGs may improve their binding to brain endothelial cells and, possibly, show subsequent transcytosis across the BBB.
Collapse
Affiliation(s)
- Bhagyashree S Joshi
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Inge S Zuhorn
- Department of Biomedical Engineering, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Li S, Wang Y, Jiang D, Ni D, Kutyreff CJ, Barnhart TE, Engle JW, Cai W. Spatiotemporal Distribution of Agrin after Intrathecal Injection and Its Protective Role in Cerebral Ischemia/Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902600. [PMID: 32076591 PMCID: PMC7029627 DOI: 10.1002/advs.201902600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/18/2019] [Indexed: 05/30/2023]
Abstract
Intrathecal injection, drugs transporting along perivascular spaces, represents an important route for maintaining blood-brain barrier (BBB) integrity after cerebral ischemia/reperfusion (I/R) injury. However, after being directly injected into cerebrospinal fluid (CSF), the temporal and spatial changes in the distribution of therapeutic protein drugs have remained unknown. Here, with positron emission tomography (PET) imaging, the uptake of 89Zr-agrin is noninvasively and dynamically monitored. These data demonstrate the time-activity curve of drugs in the brain subregions and their spatial distribution in different organs after intrathecal administration. Furthermore, agrin treatment effectively inhibits BBB disruption by reducing the loss of tight-junctional proteins. Importantly, the infarct volume is reduced; the number of apoptotic neurons is decreased; and neurological function is improved in mouse I/R injury models. Thus, intrathecal injection of agrin provides the basis for a new strategy to research and develop protein drugs for reducing the aggravation of I/R injury.
Collapse
Affiliation(s)
- Shiyong Li
- Department of RehabilitationSecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Ye Wang
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
| | - Dawei Jiang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Dalong Ni
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Christopher J. Kutyreff
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Todd E. Barnhart
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Jonathan W. Engle
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| |
Collapse
|
14
|
Brennan S, Keon M, Liu B, Su Z, Saksena NK. Panoramic Visualization of Circulating MicroRNAs Across Neurodegenerative Diseases in Humans. Mol Neurobiol 2019; 56:7380-7407. [PMID: 31037649 PMCID: PMC6815273 DOI: 10.1007/s12035-019-1615-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and dementia pose one of the greatest health challenges this century. Although these NDs have been looked at as single entities, the underlying molecular mechanisms have never been collectively visualized to date. With the advent of high-throughput genomic and proteomic technologies, we now have the opportunity to visualize these diseases in a whole new perspective, which will provide a clear understanding of the primary and secondary events vital in achieving the final resolution of these diseases guiding us to new treatment strategies to possibly treat these diseases together. We created a knowledge base of all microRNAs known to be differentially expressed in various body fluids of ND patients. We then used several bioinformatic methods to understand the functional intersections and differences between AD, PD, ALS, and MS. These results provide a unique panoramic view of possible functional intersections between AD, PD, MS, and ALS at the level of microRNA and their cognate genes and pathways, along with the entities that unify and separate them. While the microRNA signatures were apparent for each ND, the unique observation in our study was that hsa-miR-30b-5p overlapped between all four NDS, and has significant functional roles described across NDs. Furthermore, our results also show the evidence of functional convergence of miRNAs which was associated with the regulation of their cognate genes represented in pathways that included fatty acid synthesis and metabolism, ECM receptor interactions, prion diseases, and several signaling pathways critical to neuron differentiation and survival, underpinning their relevance in NDs. Envisioning this group of NDs together has allowed us to propose new ways of utilizing circulating miRNAs as biomarkers and in visualizing diverse NDs more holistically . The critical molecular insights gained through the discovery of ND-associated miRNAs, overlapping miRNAs, and the functional convergence of microRNAs on vital pathways strongly implicated in neurodegenerative processes can prove immensely valuable in the identifying new generation of biomarkers, along with the development of miRNAs into therapeutics.
Collapse
Affiliation(s)
- Samuel Brennan
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Matthew Keon
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Bing Liu
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Zheng Su
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| | - Nitin K. Saksena
- Neurodegenerative Disease section, Iggy Get Out, 19a Boundary Street, Darlinghurst NSW 2010, Sydney, Australia
| |
Collapse
|
15
|
Endothelial heparan sulfate deficiency reduces inflammation and fibrosis in murine diabetic nephropathy. J Transl Med 2018; 98:427-438. [PMID: 29330473 PMCID: PMC6247417 DOI: 10.1038/s41374-017-0015-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/22/2017] [Accepted: 12/14/2017] [Indexed: 01/08/2023] Open
Abstract
Inflammation plays a vital role in the development of diabetic nephropathy, but the underlying regulatory mechanisms are only partially understood. Our previous studies demonstrated that, during acute inflammation, endothelial heparan sulfate (HS) contributes to the adhesion and transendothelial migration of leukocytes into perivascular tissues by direct interaction with L-selectin and the presentation of bound chemokines. In the current study, we aimed to assess the role of endothelial HS on chronic renal inflammation and fibrosis in a diabetic nephropathy mouse model. To reduce sulfation of HS specifically in the endothelium, we generated Ndst1 f/f Tie2Cre + mice in which N-deacetylase/N-sulfotransferase-1 (Ndst1), the gene that initiates HS sulfation modifications in HS biosynthesis, was expressly ablated in endothelium. To induce diabetes, age-matched male Ndst1 f/f Tie2Cre - (wild type) and Ndst1 f/f Tie2Cre + mice on a C57Bl/6J background were injected intraperitoneally with streptozotocin (STZ) (50 mg/kg) on five consecutive days (N = 10-11/group). Urine and plasma were collected. Four weeks after diabetes induction the animals were sacrificed and kidneys were analyzed by immunohistochemistry and qRT-PCR. Compared to healthy controls, diabetic Ndst1 f/f Tie2Cre - mice showed increased glomerular macrophage infiltration, mannose binding lectin complement deposition and glomerulosclerosis, whereas these pathological reactions were prevented significantly in the diabetic Ndst1 f/f Tie2Cre + animals (all three p < 0.01). In addition, the expression of the podocyte damage marker desmin was significantly higher in the Ndst1 f/f Tie2Cre - group compared to the Ndst1 f/f Tie2Cre + animals (p < 0.001), although both groups had comparable numbers of podocytes. In the cortical tubulo-interstitium, similar analyses show decreased interstitial macrophage accumulation in the diabetic Ndst1 f/f Tie2Cre + animals compared to the diabetic Ndst1 f/f Tie2Cre - mice (p < 0.05). Diabetic Ndst1 f/f Tie2Cre + animals also showed reduced interstitial fibrosis as evidenced by reduced density of αSMA-positive myofibroblasts (p < 0.01), diminished collagen III deposition (p < 0.001) and reduced mRNA expression of collagen I (p < 0.001) and fibronectin (p < 0.001). Our studies indicate a pivotal role of endothelial HS in the development of renal inflammation and fibrosis in diabetic nephropathy in mice. These results suggest that HS is a possible target for therapy in diabetic nephropathy.
Collapse
|
16
|
Changyaleket B, Deliu Z, Chignalia AZ, Feinstein DL. Heparanase: Potential roles in multiple sclerosis. J Neuroimmunol 2017; 310:72-81. [PMID: 28778449 DOI: 10.1016/j.jneuroim.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 06/22/2017] [Accepted: 07/01/2017] [Indexed: 12/14/2022]
Abstract
Heparanase is a heparan sulfate degrading enzyme that cleaves heparan sulfate (HS) chains present on HS proteoglycans (HSPGs), and has been well characterized for its roles in tumor metastasis and inflammation. However, heparanase is emerging as a contributing factor in the genesis and severity of a variety of neurodegenerative diseases and conditions. This is in part due to the wide variety of HSPGs on which the presence or absence of HS moieties dictates protein function. This includes growth factors, chemokines, cytokines, as well as components of the extracellular matrix (ECM) which in turn regulate leukocyte infiltration into the CNS. Roles for heparanase in stroke, Alzheimer's disease, and glioma growth have been described; roles for heparanase in other disease such as multiple sclerosis (MS) are less well established. However, given its known roles in inflammation and leukocyte infiltration, it is likely that heparanase also contributes to MS pathology. In this review, we will briefly summarize what is known about heparanase roles in the CNS, and speculate as to its potential role in regulating disease progression in MS and its animal model EAE (experimental autoimmune encephalitis), which may justify testing of heparanase inhibitors for MS treatment.
Collapse
Affiliation(s)
| | - Zane Deliu
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
| | - Andreia Z Chignalia
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA; Jesse Brown Veteran Affairs Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
17
|
ten Cate-Hoek AJ, Henke PK, Wakefield TW. The post thrombotic syndrome: Ignore it and it will come back to bite you. Blood Rev 2015; 30:131-7. [PMID: 26462885 DOI: 10.1016/j.blre.2015.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/21/2015] [Indexed: 10/22/2022]
Abstract
Post thrombotic syndrome (PTS) is a very common chronic complication of deep venous thrombosis (DVT), as three out of ten patients with lower extremity DVT will develop PTS. The possibility to identify patients at risk is limited. Diagnosis is challenging, because there is no gold standard diagnostic method. Progress in diagnostic options may therefore change future diagnostic strategies. The better understanding of pathophysiologic processes that underlie PTS may stimulate the development of treatment modalities and improve and diversify management options. The quest for adequate preventive strategies and treatment is important because PTS has a detrimental effect on patients' quality of life and is associated with increased healthcare as well as societal costs. The problem of PTS prevention is therefore clearly relevant to patients, doctors as well as policy makers.
Collapse
Affiliation(s)
- Arina J ten Cate-Hoek
- Cardiovascular Center and Laboratory for Clinical Thrombosis and Hemostasis, Maastricht University Medical Center, Maastricht, the Netherlands.
| | - Peter K Henke
- Section of Vascular Surgery and the Jobst Vascular Research Laboratory, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| | - Thomas W Wakefield
- Section of Vascular Surgery and the Jobst Vascular Research Laboratory, Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
18
|
O'Carroll SJ, Kho DT, Wiltshire R, Nelson V, Rotimi O, Johnson R, Angel CE, Graham ES. Pro-inflammatory TNFα and IL-1β differentially regulate the inflammatory phenotype of brain microvascular endothelial cells. J Neuroinflammation 2015; 12:131. [PMID: 26152369 PMCID: PMC4506411 DOI: 10.1186/s12974-015-0346-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 06/17/2015] [Indexed: 11/28/2022] Open
Abstract
Background The vasculature of the brain is composed of endothelial cells, pericytes and astrocytic processes. The endothelial cells are the critical interface between the blood and the CNS parenchyma and are a critical component of the blood-brain barrier (BBB). These cells are innately programmed to respond to a myriad of inflammatory cytokines or other danger signals. IL-1β and TNFα are well recognised pro-inflammatory mediators, and here, we provide compelling evidence that they regulate the function and immune response profile of human cerebral microvascular endothelial cells (hCMVECs) differentially. Methods We used xCELLigence biosensor technology, which revealed global differences in the endothelial response between IL-1β and TNFα. xCELLigence is a label-free impedance-based biosensor, which is ideal for acute or long-term comparison of drug effects on cell behaviour. In addition, flow cytometry and multiplex cytokine arrays were used to show differences in the inflammatory responses from the endothelial cells. Results Extensive cytokine-secretion profiling and cell-surface immune phenotyping confirmed that the immune response of the hCMVEC to IL-1β was different to that of TNFα. Interestingly, of the 38 cytokines, chemokines and growth factors measured by cytometric bead array, the endothelial cells secreted only 13. Of importance was the observation that the majority of these cytokines were differentially regulated by either IL-1β or TNFα. Cell-surface expression of ICAM-1 and VCAM-1 were also differentially regulated by IL-1β or TNFα, where TNFα induced a substantially higher level of expression of both key leukocyte-adhesion molecules. A range of other cell-surface cellular and junctional adhesion molecules were basally expressed by the hCMVEC but were unaffected by IL-1β or TNFα. Conclusions To our knowledge, this is the most comprehensive analysis of the immunological profile of brain endothelial cells and the first direct evidence that human brain endothelial cells are differentially regulated by these two key pro-inflammatory mediators. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0346-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simon J O'Carroll
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. .,Department of Anatomy, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Dan Ting Kho
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Rachael Wiltshire
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Vicky Nelson
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Odunayo Rotimi
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. .,Department of Anatomy, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Rebecca Johnson
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Catherine E Angel
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand.
| | - E Scott Graham
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. .,Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
19
|
Abstract
UNLABELLED Nipah virus and Hendra virus are emerging, highly pathogenic, zoonotic paramyxoviruses that belong to the genus Henipavirus. They infect humans as well as numerous mammalian species. Both viruses use ephrin-B2 and -B3 as cell entry receptors, and following initial entry into an organism, they are capable of rapid spread throughout the host. We have previously reported that Nipah virus can use another attachment receptor, different from its entry receptors, to bind to nonpermissive circulating leukocytes, thereby promoting viral dissemination within the host. Here, this attachment molecule was identified as heparan sulfate for both Nipah virus and Hendra virus. Cells devoid of heparan sulfate were not able to mediate henipavirus trans-infection and showed reduced permissivity to infection. Virus pseudotyped with Nipah virus glycoproteins bound heparan sulfate and heparin but no other glycosaminoglycans in a surface plasmon resonance assay. Furthermore, heparin was able to inhibit the interaction of the viruses with the heparan sulfate and to block cell-mediated trans-infection of henipaviruses. Moreover, heparin was shown to bind to ephrin-B3 and to restrain infection of permissive cells in vitro. Consequently, treatment with heparin devoid of anticoagulant activity improved the survival of Nipah virus-infected hamsters. Altogether, these results reveal heparan sulfate as a new attachment receptor for henipaviruses and as a potential therapeutic target for the development of novel approaches against these highly lethal infections. IMPORTANCE The Henipavirus genus includes two closely related, highly pathogenic paramyxoviruses, Nipah virus and Hendra virus, which cause elevated morbidity and mortality in animals and humans. Pathogenesis of both Nipah virus and Hendra virus infection is poorly understood, and efficient antiviral treatment is still missing. Here, we identified heparan sulfate as a novel attachment receptor used by both viruses to bind host cells. We demonstrate that heparin was able to inhibit the interaction of the viruses with heparan sulfate and to block cell-mediated trans-infection of henipaviruses. Moreover, heparin also bound to the viral entry receptor and thereby restricted infection of permissive cells in vitro. Consequently, heparin treatment improved survival of Nipah virus-infected hamsters. These results uncover an important role of heparan sulfate in henipavirus infection and open novel perspectives for the development of heparan sulfate-targeting therapeutic approaches for these emerging infections.
Collapse
|
20
|
Towards understanding the roles of heparan sulfate proteoglycans in Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:516028. [PMID: 25157361 PMCID: PMC4135094 DOI: 10.1155/2014/516028] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 07/12/2014] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive loss of memory and cognitive dysfunctions. A central pathological event of AD is accumulation and deposition of cytotoxic amyloid-β peptide (Aβ) in the brain parenchyma. Heparan sulfate proteoglycans (HSPGs) and the side chains heparan sulfate (HS) are found associated with Aβ deposits in the brains of AD patients and transgenic animal models of AD. A growing body of evidence from in vitro and in vivo studies suggests functional roles of HSPG/HS in Aβ pathogenesis. Although the question of "how and why HSPG/HS is codeposited with Aβ?" still remains, it is within reach to understand the mechanisms of the events. Recent progress by immunohistochemical examination with advanced antibodies shed light on molecular structures of HS codeposited with Aβ. Several recent reports have provided important new insights into the roles of HSPG in Aβ pathogenesis. Particularly, experiments on mouse models revealed indispensible functions of HSPG in modulating Aβ-associated neuroinflammation and clearance of Aβ from the brain. Application of molecules to interfere with the interaction between HS and Aβ peptides has demonstrated beneficial effects on AD mouse models. Elucidating the functions of HSPG/HS in Aβ deposition and toxicity is leading to further understanding of the complex pathology of AD. The progress is encouraging development of new treatments for AD by targeting HS-Aβ interactions.
Collapse
|
21
|
Ge XN, Ha SG, Rao A, Greenberg YG, Rushdi MN, Esko JD, Rao SP, Sriramarao P. Endothelial and leukocyte heparan sulfates regulate the development of allergen-induced airway remodeling in a mouse model. Glycobiology 2014; 24:715-27. [PMID: 24794009 DOI: 10.1093/glycob/cwu035] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Heparan sulfate (HS) proteoglycans (HSPGs) participate in several aspects of inflammation because of their ability to bind to growth factors, chemokines, interleukins and extracellular matrix proteins as well as promote inflammatory cell trafficking and migration. We investigated whether HSPGs play a role in the development of airway remodeling during chronic allergic asthma using mice deficient in endothelial- and leukocyte-expressed N-deacetylase/N-sulfotransferase-1 (Ndst1), an enzyme involved in modification reactions during HS biosynthesis. Ndst1-deficient and wild-type (WT) mice exposed to repetitive allergen (ovalbumin [OVA]) challenge were evaluated for the development of airway remodeling. Chronic OVA-challenged WT mice exhibited increased HS expression in the lungs along with airway eosinophilia, mucus hypersecretion, peribronchial fibrosis, increased airway epithelial thickness and smooth muscle mass. In OVA-challenged Ndst1-deficient mice, lung eosinophil and macrophage infiltration as well as airway mucus accumulation, peribronchial fibrosis and airway epithelial thickness were significantly lower than in allergen-challenged WT mice along with a trend toward decreased airway smooth muscle mass. Leukocyte and endothelial Ndst 1 deficiency also resulted in significantly decreased expression of IL-13 as well as remodeling-associated mediators such as VEGF, FGF-2 and TGF-β1 in the lung tissue. At a cellular level, exposure to eotaxin-1 failed to induce TGF-β1 expression by Ndst1-deficient eosinophils relative to WT eosinophils. These studies suggest that leukocyte and endothelial Ndst1-modified HS contribute to the development of allergen-induced airway remodeling by promoting recruitment of inflammatory cells as well as regulating expression of pro-remodeling factors such as IL-13, VEGF, TGF-β1 and FGF-2 in the lung.
Collapse
Affiliation(s)
- Xiao Na Ge
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Sung Gil Ha
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Amrita Rao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Yana G Greenberg
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Muaz Nik Rushdi
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Jeffrey D Esko
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Savita P Rao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - P Sriramarao
- Department of Veterinary & Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
22
|
Zhang X, Wang B, Li JP. Implications of heparan sulfate and heparanase in neuroinflammation. Matrix Biol 2014; 35:174-81. [PMID: 24398134 DOI: 10.1016/j.matbio.2013.12.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/23/2013] [Accepted: 12/23/2013] [Indexed: 12/24/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs), expressed on the cell surface and in the extracellular matrix of most animal tissues, have essential functions in development and homeostasis, and have been implicated in several pathological conditions. The functions of HSPGs are mainly mediated through interactions of the heparan sulfate (HS) polysaccharide side chains with different protein ligands. The molecular structure of HS is highly diverse, expressed in a cell-type specific manner. The flexible yet controlled structure of HS is primarily generated through a strictly regulated biosynthesis process and is further modified post-synthetically, such as desulfation by endosulfatases and fragmentation by heparanase. Heparanase is an endo-glucuronidase expressed in all tissues. The enzyme has been found up-regulated in a number of pathological conditions, implying a role in diseases mainly through degradation of HS. Emerging evidence demonstrates important roles of HS and heparanase in inflammatory reactions, particularly in the regulation of leukocyte activation and extravasation. Neuroinflammation is a common feature of various central nervous system disorders, thus it is a great interest to understand the implications of HS and heparanase in neuroinflammation.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Neuroscience Pharmacology, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - Bo Wang
- Department of Neuropharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Center, Uppsala, Sweden.
| |
Collapse
|
23
|
Lee S, Eskin SG, Shah AK, Schildmeyer LA, McIntire LV. Effect of zinc and nitric oxide on monocyte adhesion to endothelial cells under shear stress. Ann Biomed Eng 2011; 40:697-706. [PMID: 22009315 DOI: 10.1007/s10439-011-0434-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 10/06/2011] [Indexed: 11/26/2022]
Abstract
This study describes the effect of zinc on monocyte adhesion to endothelial cells under different shear stress regimens, which may trigger atherogenesis. Human umbilical vein endothelial cells were exposed to steady shear stress (15 dynes/cm(2) or 1 dyne/cm(2)) or reversing shear stress (time average 1 dyne/cm(2)) for 24 h. In all shear stress regimes, zinc deficiency enhanced THP-1 cell adhesion, while heparinase III reduced monocyte adhesion following reversing shear stress exposure. Unlike other shear stress regimes, reversing shear stress alone enhanced monocyte adhesion, which may be associated with increased H(2)O(2) and superoxide together with relatively low levels of nitric oxide (NO) production. L-N(G)-Nitroarginine methyl ester (L-NAME) treatment increased monocyte adhesion under 15 dynes/cm(2) and under reversing shear stress. After reversing shear stress, monocyte adhesion dramatically increased with heparinase III treatment followed by a zinc scavenger. Static culture experiments supported the reduction of monocyte adhesion by zinc following endothelial cell cytokine activation. These results suggest that endothelial cell zinc levels are important for the inhibition of monocyte adhesion to endothelial cells, and may be one of the key factors in the early stages of atherogenesis.
Collapse
Affiliation(s)
- Sungmun Lee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University School of Medicine, Atlanta, GA 30332-0535, USA
| | | | | | | | | |
Collapse
|
24
|
Glycosaminoglycan binding facilitates entry of a bacterial pathogen into central nervous systems. PLoS Pathog 2011; 7:e1002082. [PMID: 21731486 PMCID: PMC3121876 DOI: 10.1371/journal.ppat.1002082] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Accepted: 04/11/2011] [Indexed: 11/26/2022] Open
Abstract
Certain microbes invade brain microvascular endothelial cells (BMECs) to breach the blood-brain barrier (BBB) and establish central nervous system (CNS) infection. Here we use the leading meningitis pathogen group B Streptococcus (GBS) together with insect and mammalian infection models to probe a potential role of glycosaminoglycan (GAG) interactions in the pathogenesis of CNS entry. Site-directed mutagenesis of a GAG-binding domain of the surface GBS alpha C protein impeded GBS penetration of the Drosophila BBB in vivo and diminished GBS adherence to and invasion of human BMECs in vitro. Conversely, genetic impairment of GAG expression in flies or mice reduced GBS dissemination into the brain. These complementary approaches identify a role for bacterial-GAG interactions in the pathogenesis of CNS infection. Our results also highlight how the simpler yet genetically conserved Drosophila GAG pathways can provide a model organism to screen candidate molecules that can interrupt pathogen-GAG interactions for future therapeutic applications. Streptococcus agalactiae (Group B Streptococcus, GBS) is a leading cause of meningitis in human newborn infants. The bacterial and host factors that allow this pathogen to cross the blood-brain barrier (BBB) and cause central nervous system (CNS) infection are not well understood. Here we demonstrate that GBS expresses a specific protein on its surface that can bind to sugar molecules known as glycosaminoglycans (GAGs) on the surface of brain capillary cells, initiating infection of the BBB. Fruit flies or mice genetically engineered to have reduced GAGs showed decreased dissemination of GBS into the brain tissues following experimental infection. Our results identify a role for bacterial-GAG interactions in the pathogenesis of newborn meningitis and highlight how the simpler yet genetically conserved fruit fly GAG biosynthetic pathways make the fruit fly a good model organism to screen candidate molecules that can interrupt pathogen-GAG interactions for future therapeutic applications.
Collapse
|
25
|
Leonardo CC, Pennypacker KR. Neuroinflammation and MMPs: potential therapeutic targets in neonatal hypoxic-ischemic injury. J Neuroinflammation 2009; 6:13. [PMID: 19368723 PMCID: PMC2674036 DOI: 10.1186/1742-2094-6-13] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Accepted: 04/15/2009] [Indexed: 12/18/2022] Open
Abstract
Exposure to hypoxic-ischemic insults during the neonatal or perinatal developmental periods produces various forms of pathology. Injuries that occur in response to these events often manifest as severe cognitive and/or motor disturbances over time. Due to difficulties regarding the early diagnosis and treatment of hypoxic-ischemic injury, there is a growing need for effective therapies that can be delivered at delayed time points. Much of the research into mechanisms of neural injury has focused on molecular targets associated with excitotoxicity and free oxygen radicals. Despite repeated success in animal models, these compounds have failed to show efficacy in clinical trials. Increasing evidence indicates that hypoxic-ischemic injury in the neonate is progressive, and the resulting neuropathies are linked to the activation of neuroinflammatory processes that occur in response to the initial wave of cell death. Understanding this latter response, therefore, will be critical in the development of novel therapies to block the progression of the injury. In this review, we summarize emerging concepts from rodent models concerning the regulation of various cytokines, chemokines, and matrix metalloproteinases in response to ischemia, and the various ways in which the delayed neuroinflammatory response may contribute to the progressive nature of neonatal hypoxic-ischemic injury in rat. Finally, we discuss data that supports the potential to target these neuroinflammatory signals at clinically relevant time points.
Collapse
Affiliation(s)
- Christopher C Leonardo
- Department of Molecular Pharmacology and Physiology, School of Basic Biomedical Sciences, College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | | |
Collapse
|
26
|
Buhler LA, Samara R, Guzman E, Wilson CL, Krizanac-Bengez L, Janigro D, Ethell DW. Matrix metalloproteinase-7 facilitates immune access to the CNS in experimental autoimmune encephalomyelitis. BMC Neurosci 2009; 10:17. [PMID: 19267908 PMCID: PMC2660336 DOI: 10.1186/1471-2202-10-17] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Accepted: 03/06/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Metalloproteinase inhibitors can protect mice against experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). Matrix metalloproteinase-9 (MMP-9) has been implicated, but it is not clear if other MMPs are also involved, including matrilysin/MMP-7 - an enzyme capable of cleaving proteins that are essential for blood brain barrier integrity and immune suppression. RESULTS Here we report that MMP-7-deficient (mmp7-/-) mice on the C57Bl/6 background are resistant to EAE induced by myelin oligodendrocyte glycoprotein (MOG). Brain sections from MOG-primed mmp7-/-mice did not show signs of immune cell infiltration of the CNS, but MOG-primed wild-type mice showed extensive vascular cuffing and mononuclear cell infiltration 15 days after vaccination. At the peak of EAE wild-type mice had MMP-7 immuno-reactive cells in vascular cuffs that also expressed the macrophage markers Iba-1 and Gr-1, as well as tomato lectin. MOG-specific proliferation of splenocytes, lymphocytes, CD4+ and CD8+ cells were reduced in cells isolated from MOG-primed mmp7-/- mice, compared with MOG-primed wild-type mice. However, the adoptive transfer of splenocytes and lymphocytes from MOG-primed mmp7-/- mice induced EAE in naïve wild-type recipients, but not naïve mmp7-/- recipients. Finally, we found that recombinant MMP-7 increased permeability between endothelial cells in an in vitro blood-brain barrier model. CONCLUSION Our findings suggest that MMP-7 may facilitate immune cell access or re-stimulation in perivascular areas, which are critical events in EAE and multiple sclerosis, and provide a new therapeutic target to treat this disorder.
Collapse
Affiliation(s)
- Lillian A Buhler
- Division of Biomedical Sciences, University of California Riverside, 900 University Avenue, Riverside, CA 92521-0121, USA
- Biochemistry and Molecular Biology Graduate Program, UCR, Riverside, CA 92521, USA
| | - Ramsey Samara
- Division of Biomedical Sciences, University of California Riverside, 900 University Avenue, Riverside, CA 92521-0121, USA
- Neuroscience Graduate Program, UCR, Riverside, CA 92521, USA
| | - Esther Guzman
- Division of Biomedical Sciences, University of California Riverside, 900 University Avenue, Riverside, CA 92521-0121, USA
| | - Carole L Wilson
- Department of Pathology, University of Washington School of Medicine, 300 9th Avenue, Seattle, WA 98104, USA
| | - Liljana Krizanac-Bengez
- Cerebrovascular Research, Cleveland Clinic Foundation NB20, Neurosurgery, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Damir Janigro
- Cerebrovascular Research, Cleveland Clinic Foundation NB20, Neurosurgery, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Douglas W Ethell
- Division of Biomedical Sciences, University of California Riverside, 900 University Avenue, Riverside, CA 92521-0121, USA
| |
Collapse
|
27
|
Alteration of blood-brain barrier integrity by retroviral infection. PLoS Pathog 2008; 4:e1000205. [PMID: 19008946 PMCID: PMC2575404 DOI: 10.1371/journal.ppat.1000205] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2008] [Accepted: 10/15/2008] [Indexed: 01/18/2023] Open
Abstract
The blood–brain barrier (BBB), which forms the interface between the blood and the cerebral parenchyma, has been shown to be disrupted during retroviral-associated neuromyelopathies. Human T Lymphotropic Virus (HTLV-1) Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP) is a slowly progressive neurodegenerative disease associated with BBB breakdown. The BBB is composed of three cell types: endothelial cells, pericytes and astrocytes. Although astrocytes have been shown to be infected by HTLV-1, until now, little was known about the susceptibility of BBB endothelial cells to HTLV-1 infection and the impact of such an infection on BBB function. We first demonstrated that human cerebral endothelial cells express the receptors for HTLV-1 (GLUT-1, Neuropilin-1 and heparan sulfate proteoglycans), both in vitro, in a human cerebral endothelial cell line, and ex vivo, on spinal cord autopsy sections from HAM/TSP and non-infected control cases. In situ hybridization revealed HTLV-1 transcripts associated with the vasculature in HAM/TSP. We were able to confirm that the endothelial cells could be productively infected in vitro by HTLV-1 and that blocking of either HSPGs, Neuropilin 1 or Glut1 inhibits this process. The expression of the tight-junction proteins within the HTLV-1 infected endothelial cells was altered. These cells were no longer able to form a functional barrier, since BBB permeability and lymphocyte passage through the monolayer of endothelial cells were increased. This work constitutes the first report of susceptibility of human cerebral endothelial cells to HTLV-1 infection, with implications for HTLV-1 passage through the BBB and subsequent deregulation of the central nervous system homeostasis. We propose that the susceptibility of cerebral endothelial cells to retroviral infection and subsequent BBB dysfunction is an important aspect of HAM/TSP pathogenesis and should be considered in the design of future therapeutics strategies. The blood–brain barrier (BBB) forms the interface between the blood and the central nervous system (CNS). BBB disruption is considered to be a key event in the pathogenesis of retroviral-associated neurological diseases. The present paper deals with the susceptibility of the endothelial cells (i.e., one of the main cellular components of BBB) to retroviral infection, and with the impact of infection in BBB function. This study focuses on the Human T-Lymphotropic Virus (HTLV-1), which infects 20 million people worldwide, and is the etiological agent of a neurodegenerative disease called HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). We first demonstrated that the cerebral endothelial cells express the receptors for the retrovirus in vitro, and on spinal cord autopsy sections from non-infected and HAM/TSP patients. We found on these latter that vascular-like structures were infected and confirmed in vitro that the endothelial cells could be productively infected by HTLV-1. We demonstrated that such an infection impairs BBB properties in vitro, as well as tight junctions, that are cell adhesion structures. This study is the first to demonstrate the impact of HTLV-1 infection on human BBB integrity; such a susceptibility has to be considered in the design of future therapeutics strategies.
Collapse
|
28
|
Reijerkerk A, Kooij G, van der Pol SMA, Leyen T, van Het Hof B, Couraud PO, Vivien D, Dijkstra CD, de Vries HE. Tissue-type plasminogen activator is a regulator of monocyte diapedesis through the brain endothelial barrier. THE JOURNAL OF IMMUNOLOGY 2008; 181:3567-74. [PMID: 18714030 DOI: 10.4049/jimmunol.181.5.3567] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inflammatory cell trafficking into the brain complicates several neurological disorders including multiple sclerosis. Normally, reliable brain functioning is maintained and controlled by the blood-brain barrier (BBB), which is essential to restrict the entry of potentially harmful molecules and cells from the blood into the brain. The BBB is a selective barrier formed by dedicated brain endothelial cells and dependent on the presence of intracellular tight junctions. In multiple sclerosis, a severe dysfunction of the BBB is observed, which is key to monocyte infiltration and inflammation in the brain. Proteolytic activity has been associated with these inflammatory processes in the brain. Our studies in plasma of rats indicated that the extracellular protease tissue-type plasminogen activator (tPA) correlates with the clinical signs of experimental allergic encephalomyelitis, a rat model of multiple sclerosis. In this study, we studied the function of the tPA during diapedesis of monocytes through a rat and human brain endothelial barrier. Monocyte-brain endothelial cell coculture experiments showed that monocytes induce the release of tPA by brain endothelial cells, which subsequently activates the signal transduction protein extracellular signal related kinase (ERK1/2), both involved in monocyte diapedesis. Importantly, live imaging and immunoblot analyses of rat brain endothelial cells revealed that tPA and ERK1/2 control the breakdown of the tight junction protein occludin. These studies identify tPA as a novel and relevant pathological mediator of neuroinflammation and provide a potential mechanism for this.
Collapse
Affiliation(s)
- Arie Reijerkerk
- Neuroimmunology Research Group, Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Patterson AM, Cartwright A, David G, Fitzgerald O, Bresnihan B, Ashton BA, Middleton J. Differential expression of syndecans and glypicans in chronically inflamed synovium. Ann Rheum Dis 2008; 67:592-601. [PMID: 17545191 PMCID: PMC2563418 DOI: 10.1136/ard.2006.063875] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2007] [Indexed: 12/14/2022]
Abstract
BACKGROUND Membrane-bound heparan sulphate proteoglycans (HSPGs) act as co-receptors and presenters of cytokines and are involved in cell-matrix and cell-cell adhesion. AIM To investigate which HSPGs are expressed in knee joint synovia from patients with different forms of arthritis and normal individuals. METHODS Synovial samples were obtained from patients with early rheumatoid arthritis (n = 8), longstanding rheumatoid arthritis (n = 13), psoriatic arthritis (n = 7), osteoarthritis (n = 6) and normal joints (n = 12). Expression of syndecan-1, -2, -3 and -4 and glypican-1, -3 and -4 was analysed by immunohistochemistry and dual label immunofluorescence. RESULTS The expression of HSPGs in chronically inflamed synovium exhibited a differential distribution. Syndecan-1 was present in the mononuclear infiltrates of synovia from patients with rheumatoid and psoriatic arthritis where it was expressed by plasma cells. Syndecan-2 was present mainly in blood vessels where it occurred on endothelial cells, pericytes and smooth muscle cells. Syndecan-3 stained intensely in endothelial cells but also occurred in sublining macrophages and the lining layer. Glypican-4 occurred in the lining layer and blood vessels. Increased expression of these HSPGs was apparent in rheumatoid and psoriatic compared to osteoarthritic and normal synovia. Little or no staining for syndecan-4, glypican-1 and glypican-3 was seen in all samples. DISCUSSION Selected HSPGs, such as syndecan-1, -2 and -3 and glypican-4, could play a part in the pathophysiology of arthritis, such as the migration and retention of leukocytes and angiogenesis in the chronically inflamed synovium.
Collapse
Affiliation(s)
- A M Patterson
- Leopold Muller Arthritis Research Centre, Institute for Science and Technology in Medicine, Medical School, Keele University at Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - A Cartwright
- Leopold Muller Arthritis Research Centre, Institute for Science and Technology in Medicine, Medical School, Keele University at Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - G David
- Center for Human Genetics, University of Leuven, Campus Gasthuisberg, Leuven, Belgium
| | - O Fitzgerald
- University Department of Rheumatology, St Vincents Hospital, Dublin, Ireland
| | - B Bresnihan
- University Department of Rheumatology, St Vincents Hospital, Dublin, Ireland
| | - B A Ashton
- Leopold Muller Arthritis Research Centre, Institute for Science and Technology in Medicine, Medical School, Keele University at Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - J Middleton
- Leopold Muller Arthritis Research Centre, Institute for Science and Technology in Medicine, Medical School, Keele University at Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| |
Collapse
|
30
|
Interaction of inflammation, thrombosis, aspirin and enoxaparin in CNS experimental antiphospholipid syndrome. Neurobiol Dis 2008; 30:56-64. [DOI: 10.1016/j.nbd.2007.12.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 12/05/2007] [Accepted: 12/10/2007] [Indexed: 11/22/2022] Open
|
31
|
Sha G, Wu D, Zhang L, Chen X, Lei M, Sun H, Lin S, Lang J. Differentially expressed genes in human endometrial endothelial cells derived from eutopic endometrium of patients with endometriosis compared with those from patients without endometriosis. Hum Reprod 2007; 22:3159-69. [DOI: 10.1093/humrep/dem266] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
32
|
Schreibelt G, Musters RJP, Reijerkerk A, de Groot LR, van der Pol SMA, Hendrikx EML, Döpp ED, Dijkstra CD, Drukarch B, de Vries HE. Lipoic Acid Affects Cellular Migration into the Central Nervous System and Stabilizes Blood-Brain Barrier Integrity. THE JOURNAL OF IMMUNOLOGY 2006; 177:2630-7. [PMID: 16888025 DOI: 10.4049/jimmunol.177.4.2630] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Reactive oxygen species (ROS) play an important role in various events underlying multiple sclerosis (MS) pathology. In the initial phase of lesion formation, ROS are known to mediate the transendothelial migration of monocytes and induce a dysfunction of the blood-brain barrier (BBB). In this study, we describe the beneficial effect of the antioxidant alpha-lipoic acid (LA) on these phenomena. In vivo, LA dose-dependently prevented the development of clinical signs in a rat model for MS, acute experimental allergic encephalomyelitis (EAE). Clinical improvement was coupled to a decrease in leukocyte infiltration into the CNS, in particular monocytes. Monocytes isolated from the circulation of LA-treated rats revealed a reduced migratory capacity to cross a monolayer of rat brain endothelial cells in vitro compared with monocytes isolated from untreated EAE controls. Using live cell imaging techniques, we visualized and quantitatively assessed that ROS are produced within minutes upon the interaction of monocytes with brain endothelium. Monocyte adhesion to an in vitro model of the BBB subsequently induced enhanced permeability, which could be inhibited by LA. Moreover, administration of exogenous ROS to brain endothelial cells induced cytoskeletal rearrangements, which was inhibited by LA. In conclusion, we show that LA has a protective effect on EAE development not only by affecting the migratory capacity of monocytes, but also by stabilization of the BBB, making LA an attractive therapeutic agent for the treatment of MS.
Collapse
Affiliation(s)
- Gerty Schreibelt
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Wegrowski Y, Milard AL, Kotlarz G, Toulmonde E, Maquart FX, Bernard J. Cell surface proteoglycan expression during maturation of human monocytes-derived dendritic cells and macrophages. Clin Exp Immunol 2006; 144:485-93. [PMID: 16734618 PMCID: PMC1941969 DOI: 10.1111/j.1365-2249.2006.03059.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Cell surface proteoglycans play an important part in the functional and metabolic behaviour of leucocytes. We studied the expression of cell surface proteoglycans in human monocytes, in monocyte-derived immature and mature dendritic cells and in macrophages by metabolic labelling with [(35)S]-sulphate, reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting. Immature dendritic cells had the highest metabolic activity for the synthesis of cell surface proteoglycans. The major part of these proteoglycans was in phosphatidylinositol-anchored form and was released after treatment with phospholipase C. A minor part was released by trypsin. Digestion with chondroitinase ABC and mild HNO(2) treatment showed that cell surface proteoglycans had a higher proportion of chondroitin sulphate, both in the phospholipase C and trypsin fractions, suggesting that at least some glypicans contained chondroitin sulphate chains. RT-PCR detected the transcripts of glypicans 1, 3, 4 and 5 and all syndecans. Immature dendritic cells expressed a most complex spectrum of glypicans and syndecans, glypican-1 and syndecan-1 being expressed preferentially by this type of cells. Mature dendritic cells expressed glypican-3, which was not present in other lineages. These results suggest that different mononuclear cells synthesize cell surface proteoglycans actively with characteristic expression of different syndecans and glypicans genes, depending on the degree of cell differentiation and/or maturation.
Collapse
Affiliation(s)
- Y Wegrowski
- Laboratory of Biochemistry, CNRS UMR 6198, Faculty of Medicine, Reims, France.
| | | | | | | | | | | |
Collapse
|
34
|
Abulrob A, Sprong H, Van Bergen en Henegouwen P, Stanimirovic D. The blood-brain barrier transmigrating single domain antibody: mechanisms of transport and antigenic epitopes in human brain endothelial cells. J Neurochem 2006; 95:1201-14. [PMID: 16271053 DOI: 10.1111/j.1471-4159.2005.03463.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Antibodies against receptors that undergo transcytosis across the blood-brain barrier (BBB) have been used as vectors to target drugs or therapeutic peptides into the brain. We have recently discovered a novel single domain antibody, FC5, which transmigrates across human cerebral endothelial cells in vitro and the BBB in vivo. The purpose of this study was to characterize mechanisms of FC5 endocytosis and transcytosis across the BBB and its putative receptor on human brain endothelial cells. The transport of FC5 across human brain endothelial cells was polarized, charge independent and temperature dependent, suggesting a receptor-mediated process. FC5 taken up by human brain endothelial cells co-localized with clathrin but not with caveolin-1 by immunochemistry and was detected in clathrin-enriched subcellular fractions by western blot. The transendothelial migration of FC5 was reduced by inhibitors of clathrin-mediated endocytosis, K+ depletion and chlorpromazine, but was insensitive to caveolae inhibitors, filipin, nystatin or methyl-beta-cyclodextrin. Following internalization, FC5 was targeted to early endosomes, bypassed late endosomes/lysosomes and remained intact after transcytosis. The transcytosis process was inhibited by agents that affect actin cytoskeleton or intracellular signaling through PI3-kinase. Pretreatment of human brain endothelial cells with wheatgerm agglutinin, sialic acid, alpha(2,3)-neuraminidase or Maackia amurensis agglutinin that recognizes alpha(2,3)-, but not with Sambucus nigra agglutinin that recognizes alpha(2,6) sialylgalactosyl residues, significantly reduced FC5 transcytosis. FC5 failed to recognize brain endothelial cells-derived lipids, suggesting that it binds luminal alpha(2,3)-sialoglycoprotein receptor which triggers clathrin-mediated endocytosis. This putative receptor may be a new target for developing brain-targeting drug delivery vectors.
Collapse
Affiliation(s)
- Abedelnasser Abulrob
- Cerebrovascular Research Group, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
35
|
Briani C, Cagnin A, Gallo L, Toffanin E, Varagnolo M, Zaninotto M, Plebani M, Ricchieri G, Battistin L, Pizzolato G. Anti-heparan sulphate antibodies and homocysteine in dementia: markers of vascular pathology? J Neurol Sci 2005; 229-230:215-8. [PMID: 15760642 DOI: 10.1016/j.jns.2004.11.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Increasing evidence supports a pathogenic role of heparan sulphate (HS) in the development of dementia. Since HS proteoglycans are present in the endothelial cells and perivascular basement membrane, we wanted to assess blood titres of HS antibodies (Abs) in patients with vascular dementia (VD) and in patients with Alzheimer's disease (AD) with cerebrovascular disease (CVD) [mixed dementia (MixD)]. Moreover, plasma levels of homocysteine, an independent risk factor for the development of dementia as well as for CVD, were also determined. High HS Abs titres were present in one patient with VD and in two patients with mixed dementia, as well as in two neurological control patients (stroke and epilepsy). Increased homocysteine levels were found in 62.5% of patients with mixed dementia, in 22.2% of the VD subjects, in 54.2% of patients with CVD, and in 41.2% of patients with other neurological diseases. The present findings suggest that neither elevated HS Abs titres nor increased homocysteinemia may represent a useful biochemical marker for the diagnosis of VD.
Collapse
Affiliation(s)
- Chiara Briani
- Department of Neurosciences, University of Padova, Via Giustiniani, 5, 35128 Padova, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kumar A, Kingdon E, Norman J. The isoprostane 8-iso-PGF2alpha suppresses monocyte adhesion to human microvascular endothelial cells via two independent mechanisms. FASEB J 2005; 19:443-5. [PMID: 15640282 DOI: 10.1096/fj.03-1364fje] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Isoprostanes, produced in vivo by non-enzymatic free-radical-induced lipid peroxidation, are markers of oxidative stress. Elevated serum and urine levels of 8-iso-PGF2alpha have been reported in a variety of diseases, many of which are characterized by early perivascular inflammatory infiltrates. It has been suggested that, in addition to being markers of oxidative stress, isoprostanes may have pathogenic functions. In this study, we investigated the potential role of 8-iso-PGF2alpha in inflammation, focusing on its effects on adhesion of monocytes to microvascular endothelial cells, an early event in the inflammatory response. In monocyte adhesion assays, 8-iso-PGF2alpha (>10(-8) M) suppressed both basal and TNF-alpha-induced monocyte adhesion to quiescent or proliferating human dermal (HMEC) and rat renal microvascular endothelial cells. In contrast, 8-iso-PGF2alpha stimulated monocyte adhesion to human umbilical vein endothelial cells (HUVEC) as also reported by others. 8-Iso-PGF2alpha had no effect on the viability (Trypan Blue exclusion) of U937 monocytes or HMEC. 8-Iso-PGF2alpha also had no effect on HMEC surface expression of ICAM-1 or VCAM-1. Exposure of HMEC to 8-iso-PGF2alpha for 1-2 h was sufficient to reduce monocyte adhesion to the cell surface, and this effect was independent of de novo protein synthesis by HMEC. The effect of 8-iso-PGF2alpha was mimicked by a thromboxane receptor (TP) agonist (U46619) and blocked by a TP antagonist (SQ29548), indicating a TP-mediated process. Signal transduction pathway inhibitors (SB203580, curcumin, and PD98059) implicated p38 and JNK, but not ERK, in 8-iso-PGF2alpha-induced suppression of monocyte adhesion. In addition to a direct effect, conditioned medium (CM) transfer experiments suggest that 8-iso-PGF2alpha induces a secondary mediator, which also suppresses monocyte adhesion but via an alternative mechanism initiated between 3-4 h, which is TP-independent, requires new protein synthesis, and is primarily dependent on activation of p38. The data show that 8-iso-PGF2alpha can suppress the attachment of monocytes to HMECs via two independent pathways, indicating a potential anti-inflammatory effect of 8-iso-PGF2alpha in the microvasculature.
Collapse
MESH Headings
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
- Animals
- Bridged Bicyclo Compounds, Heterocyclic
- Cell Adhesion/drug effects
- Cell Line
- Culture Media, Conditioned
- Dinoprost/analogs & derivatives
- Dinoprost/pharmacology
- Dinoprost/physiology
- Dose-Response Relationship, Drug
- Endothelial Cells/chemistry
- Endothelial Cells/physiology
- Fatty Acids, Unsaturated
- Humans
- Hydrazines/pharmacology
- Inflammation/pathology
- Intercellular Adhesion Molecule-1/analysis
- JNK Mitogen-Activated Protein Kinases/metabolism
- Kidney/blood supply
- MAP Kinase Kinase 4
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Monocytes/physiology
- Protein Synthesis Inhibitors/pharmacology
- Rats
- Receptors, Thromboxane A2, Prostaglandin H2/agonists
- Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors
- Receptors, Thromboxane A2, Prostaglandin H2/physiology
- Signal Transduction
- Skin/blood supply
- Tumor Necrosis Factor-alpha/pharmacology
- U937 Cells
- Umbilical Veins
- Vascular Cell Adhesion Molecule-1/analysis
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Anila Kumar
- Centre for Nephrology, Division of Medicine, Royal Free and University College Medical School, London, UK
| | | | | |
Collapse
|
37
|
Bobardt MD, Salmon P, Wang L, Esko JD, Gabuzda D, Fiala M, Trono D, Van der Schueren B, David G, Gallay PA. Contribution of proteoglycans to human immunodeficiency virus type 1 brain invasion. J Virol 2004; 78:6567-84. [PMID: 15163749 PMCID: PMC416544 DOI: 10.1128/jvi.78.12.6567-6584.2004] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
As a neurotropic virus, human immunodeficiency virus type 1 (HIV-1) invades the brain and causes severe neuronal, astrocyte, and myelin damage in AIDS patients. To gain access to the brain, HIV-1 must migrate through brain microvascular endothelial cells (BMECs), which compose the blood-brain barrier (BBB). Given that BMECs lack the entry receptor CD4, HIV-1 must use receptors distinct from CD4 to enter these cells. We previously reported that cell surface proteoglycans serve as major HIV-1 receptors on primary human endothelial cells. In this study, we examined whether proteoglycans also impact cell-free HIV-1 invasion of the brain. Using an artificial BBB transmigration assay, we found that both heparan and chondroitin sulfate proteoglycans (HSPGs and CSPGs, respectively) are abundantly expressed on primary BMECs and promote HIV-1 attachment and entry. In contrast, the classical entry receptors, CXCR4 and CCR5, only moderately enhanced these processes. HSPGs and CSPGs captured HIV-1 in a gp120-dependent manner. However, no correlation between coreceptor usage and transmigration was identified. Furthermore, brain-derived viruses did not transmigrate more efficiently than lymphoid-derived viruses, suggesting that the ability of HIV-1 to replicate in the brain does not correlate with its capacity to migrate through the BBB as cell-free virus. Given that HIV-1-proteoglycan interactions are based on electrostatic contacts between basic residues in gp120 and sulfate groups in proteoglycans, HIV-1 may exploit these interactions to rapidly enter and migrate through the BBB to invade the brain.
Collapse
Affiliation(s)
- Michael D Bobardt
- Department of Immunology, IMM-9, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|