1
|
Torrente D, Su EJ, Citalán-Madrid AF, Schielke GP, Magaoay D, Warnock M, Stevenson T, Mann K, Lesept F, Delétage N, Blanc M, Norris EH, Vivien D, Lawrence DA. The interaction of tPA with NMDAR1 drives neuroinflammation and neurodegeneration in α-synuclein-mediated neurotoxicity. J Neuroinflammation 2025; 22:8. [PMID: 39810216 PMCID: PMC11731172 DOI: 10.1186/s12974-025-03336-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The thrombolytic protease tissue plasminogen activator (tPA) is expressed in the CNS, where it regulates diverse functions including neuronal plasticity, neuroinflammation, and blood-brain-barrier integrity. However, its role in different brain regions such as the substantia nigra (SN) is largely unexplored. In this study, we characterize tPA expression, activity, and localization in the SN using a combination of retrograde tracing and β-galactosidase tPA reporter mice. We further investigate tPA's potential role in SN pathology in an α-synuclein mouse model of Parkinson's disease (PD). To characterize the mechanism of tPA action in α-synuclein-mediated pathology in the SN and to identify possible therapeutic pathways, we performed RNA-seq analysis of the SN and used multiple transgenic mouse models. These included tPA deficient mice and two newly developed transgenic mice, a knock-in model expressing endogenous levels of proteolytically inactive tPA (tPA Ala-KI) and a second model overexpressing proteolytically inactive tPA (tPA Ala-BAC). Our findings show that striatal GABAergic neurons send tPA+ projections to dopaminergic (DA)-neurons in the SN and that tPA is released from SN-derived synaptosomes upon stimulation. We also found that tPA levels in the SN increased following α-synuclein overexpression. Importantly, tPA deficiency protects DA-neurons from degeneration, prevents behavioral deficits, and reduces microglia activation and T-cell infiltration induced by α-synuclein overexpression. RNA-seq analysis indicates that tPA in the SN is required for the upregulation of genes involved in the innate and adaptive immune responses induced by α-synuclein overexpression. Overexpression of α-synuclein in tPA Ala-KI mice, expressing only proteolytically inactive tPA, confirms that tPA-mediated neuroinflammation and neurodegeneration is independent of its proteolytic activity. Moreover, overexpression of proteolytically inactive tPA in tPA Ala-BAC mice leads to increased neuroinflammation and neurodegeneration compared to mice expressing normal levels of tPA, suggesting a tPA dose response. Finally, treatment of mice with glunomab, a neutralizing antibody that selectively blocks tPA binding to the N-methyl-D-aspartate receptor-1 (NMDAR1) without affecting NMDAR1 ion channel function, identifies the tPA interaction with NMDAR1 as necessary for tPA-mediated neuroinflammation and neurodegeneration in response to α-synuclein-mediated neurotoxicity. Thus, our data identifies a novel pathway that promotes DA-neuron degeneration and suggests a potential therapeutic intervention for PD targeting the tPA-NMDAR1 interaction.
Collapse
Affiliation(s)
- Daniel Torrente
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, USA
| | - Enming J Su
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Alí Francisco Citalán-Madrid
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Gerald P Schielke
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Daniel Magaoay
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark Warnock
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Tamara Stevenson
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Kris Mann
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA
| | - Flavie Lesept
- Lys Therapeutics, Main offices: 56 rue Saint Jean de Dieu, Lyon, 69007, France
- Lys Therapeutics, HQ: Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Nathalie Delétage
- Lys Therapeutics, Main offices: 56 rue Saint Jean de Dieu, Lyon, 69007, France
- Lys Therapeutics, HQ: Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Manuel Blanc
- Lys Therapeutics, Main offices: 56 rue Saint Jean de Dieu, Lyon, 69007, France
- Lys Therapeutics, HQ: Cyceron, Boulevard Henri Becquerel, Caen, 14000, France
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, USA
| | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders (PhIND), UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Normandie Univ, Caen, France
- Department of Clinical Research, Caen University Hospital, CHU, Caen, France
| | - Daniel A Lawrence
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-0644, USA.
| |
Collapse
|
2
|
Wada T, Gando S. Phenotypes of Disseminated Intravascular Coagulation. Thromb Haemost 2024; 124:181-191. [PMID: 37657485 PMCID: PMC10890912 DOI: 10.1055/a-2165-1142] [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: 07/30/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023]
Abstract
Two phenotypes of disseminated intravascular coagulation (DIC) are systematically reviewed. DIC is classified into thrombotic and fibrinolytic phenotypes characterized by thrombosis and hemorrhage, respectively. Major pathology of DIC with thrombotic phenotype is the activation of coagulation, insufficient anticoagulation with endothelial injury, and plasminogen activator inhibitor-1-mediated inhibition of fibrinolysis, leading to microvascular fibrin thrombosis and organ dysfunction. DIC with fibrinolytic phenotype is defined as massive thrombin generation commonly observed in any type of DIC, combined with systemic pathologic hyperfibrinogenolysis caused by underlying disorder that results in severe bleeding due to excessive plasmin formation. Three major pathomechanisms of systemic hyperfibrinogenolysis have been considered: (1) acceleration of tissue-type plasminogen activator (t-PA) release from hypoxic endothelial cells and t-PA-rich storage pools, (2) enhancement of the conversion of plasminogen to plasmin due to specific proteins and receptors that are expressed on cancer cells and endothelial cells, and (3) alternative pathways of fibrinolysis. DIC with fibrinolytic phenotype can be diagnosed by DIC diagnosis followed by the recognition of systemic pathologic hyperfibrin(ogen)olysis. Low fibrinogen levels, high fibrinogen and fibrin degradation products (FDPs), and the FDP/D-dimer ratio are important for the diagnosis of systemic pathologic hyperfibrin(ogen)olysis. Currently, evidence-based treatment strategies for DIC with fibrinolytic phenotypes are lacking. Tranexamic acid appears to be one of the few methods to be effective in the treatment of systemic pathologic hyperfibrin(ogen)olysis. International cooperation for the elucidation of pathomechanisms, establishment of diagnostic criteria, and treatment strategies for DIC with fibrinolytic phenotype are urgent issues in the field of thrombosis and hemostasis.
Collapse
Affiliation(s)
- Takeshi Wada
- Department of Anesthesiology and Critical Care Medicine, Division of Acute and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Satoshi Gando
- Department of Anesthesiology and Critical Care Medicine, Division of Acute and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
- Department of Acute and Critical Care Medicine, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| |
Collapse
|
3
|
Ding S, Chen Q, Chen H, Luo B, Li C, Wang L, Asakawa T. The Neuroprotective Role of Neuroserpin in Ischemic and Hemorrhagic Stroke. Curr Neuropharmacol 2021; 19:1367-1378. [PMID: 33032511 PMCID: PMC8719291 DOI: 10.2174/1570159x18666201008113052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/27/2020] [Accepted: 10/05/2020] [Indexed: 11/25/2022] Open
Abstract
Tissue plasminogen activator (tPA) is commonly used to treat acute ischemic stroke within an appropriate therapeutic window. Its inhibitor, neuroserpin (NSP), is reported to exhibit neuroprotective effects on stroke. This review aims to summarize, from literature, the available evidence, potential mechanisms, and knowledge limitations regarding the neuroprotective role of NSP in stroke. All the available evidence indicates that the regulation of the inflammatory response may play a key role in the mechanisms of NSP, which involve all the constituents of the neuroimmune axis. The neuroinflammatory response triggered by stroke can be reversed by NSP, with complicated mechanisms such as maintenance and reconstruction of the structure and function of the blood-brain barrier (BBB), protection of the cells in the central nervous system, and suppression of cell death in both ischemic and hemorrhagic stroke. Moreover, available evidence strongly suggests a tPA-independent mechanism is involved in NSP. However, there are many important issues that are still unclear and need further investigation, such as the effects of NSP on hemorrhagic stroke, the role of the tPA-independent neuroprotective mechanisms, and the clinical application prospects of NSP. We believe our work will be helpful to further understand the neuroprotective role of NSP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Tetsuya Asakawa
- Address correspondence to this author at the Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shennanzhong Road 3025, Shenzhen, Guangdong Province, 518033, China; Tel: + 86-755-8398-2275; Fax: + 86-755-8398-0805; E-mail:
| |
Collapse
|
4
|
Formation of nasal polyps: The roles of innate type 2 inflammation and deposition of fibrin. J Allergy Clin Immunol 2020; 145:740-750. [PMID: 32145873 DOI: 10.1016/j.jaci.2020.01.027] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/28/2022]
Abstract
Chronic rhinosinusitis (CRS) is one of the most common chronic diseases worldwide. It is a heterogeneous disease, and geographical or ethnic differences in inflammatory pattern in nasal mucosa are major issues. Tissue eosinophilia in CRS is highly associated with extensive sinus disease, recalcitrance, and a higher nasal polyp (NP) recurrence rate after surgery. The prevalence of eosinophilic CRS (ECRS) is increasing in Asian countries within the last 2 decades, and this trend appears to be occurring across the world. International consensus criteria for ECRS are required for the accurate understanding of disease pathology and precision medicine. In a multicenter large-scale epidemiological survey, the "Japanese Epidemiological Survey of Refractory Eosinophilic Chronic Rhinosinusitis study," ECRS was definitively defined when the eosinophil count in nasal mucosa is greater than or equal to 70 eosinophils/hpf (magnification, ×400), and this study proposed an algorithm that classifies CRS into 4 groups according to disease severity. The main therapeutic goal with ECRS is to eliminate or diminish the bulk of NP tissue. NPs are unique abnormal lesions that grow from the lining of the nasal and paranasal sinuses, and type 2 inflammation plays a critical role in NP development in patients with ECRS. An imbalance between protease and endogenous protease inhibitors might play a pivotal role in the initiation and exacerbation of type 2 inflammation in ECRS. Intraepithelial mast cells in NPs, showing a tryptase+, chymase- phenotype, may also enhance type 2 inflammation. Intense edema and reduced fibrosis are important histological features of eosinophilic NPs. Mucosal edema mainly consists of exuded plasma protein, and excessive fibrin deposition would be expected to contribute to the retention of proteins from capillaries and thereby perpetuate mucosal edema that may play an etiological role in NPs. Upregulation of the coagulation cascade and downregulation of fibrinolysis strongly induce abnormal fibrin deposition in nasal mucosa, and type 2 inflammation plays a central role in the imbalance of coagulation and fibrinolysis.
Collapse
|
5
|
Yoshizaki S, Kijima K, Hara M, Saito T, Tamaru T, Tanaka M, Konno DJ, Nakashima Y, Okada S. Tranexamic acid reduces heme cytotoxicity via the TLR4/TNF axis and ameliorates functional recovery after spinal cord injury. J Neuroinflammation 2019; 16:160. [PMID: 31358003 PMCID: PMC6661785 DOI: 10.1186/s12974-019-1536-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/05/2019] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a catastrophic trauma accompanied by intralesional bleeding and neuroinflammation. Recently, there is increasing interest in tranexamic acid (TXA), an anti-fibrinolytic drug, which can reduce the bleeding volume after physical trauma. However, the efficacy of TXA on the pathology of SCI remains unknown. METHODS After producing a contusion SCI at the thoracic level of mice, TXA was intraperitoneally administered and the bleeding volume in the lesion area was quantified. Tissue damage was evaluated by immunohistochemical and gene expression analyses. Since heme is one of the degraded products of red blood cells (RBCs) and damage-associated molecular pattern molecules (DAMPs), we examined the influence of heme on the pathology of SCI. Functional recovery was assessed using the open field motor score, a foot print analysis, a grid walk test, and a novel kinematic analysis system. Statistical analyses were performed using Wilcoxon's rank-sum test, Dunnett's test, and an ANOVA with the Tukey-Kramer post-hoc test. RESULTS After SCI, the intralesional bleeding volume was correlated with the heme content and the demyelinated area at the lesion site, which were significantly reduced by the administration of TXA. In the injured spinal cord, toll-like receptor 4 (TLR4), which is a DAMP receptor, was predominantly expressed in microglial cells. Heme stimulation increased TLR4 and tumor necrosis factor (TNF) expression levels in primary microglial cells in a dose-dependent manner. Similarly to the in vitro experiments, the injection of non-lysed RBCs had little pathological influence on the spinal cord, whereas the injection of lysed RBCs or heme solution significantly upregulated the TLR4 and TNF expression in microglial cells. In TXA-treated SCI mice, the decreased expressions of TLR4 and TNF were observed at the lesion sites, accompanied by a significant reduction in the number of apoptotic cells and better functional recovery in comparison to saline-treated control mice. CONCLUSION The administration of TXA ameliorated the intralesional cytotoxicity both by reducing the intralesional bleeding volume and preventing heme induction of the TLR4/TNF axis in the SCI lesion. Our findings suggest that TXA treatment may be a therapeutic option for acute-phase SCI.
Collapse
Affiliation(s)
- Shingo Yoshizaki
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
- Department of Immunology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
- Department of Immunology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Masamitsu Hara
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Takeyuki Saito
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
- Department of Immunology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Masatake Tanaka
- Department of Immunology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Dai-jiro Konno
- Department of Immunology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
- Department of Immunology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582 Japan
| |
Collapse
|
6
|
Griemert EV, Hedrich J, Hirnet T, Thal SC. Deficiency of Plasminogen Activator Inhibitor Type 2 Limits Brain Edema Formation after Traumatic Brain Injury. J Neurotrauma 2019; 36:2272-2278. [DOI: 10.1089/neu.2018.6126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Eva-Verena Griemert
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | - Jana Hedrich
- Institute of Physiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
- Max Planck Institute for Polymer Research, Mainz, Germany
| | - Tobias Hirnet
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | - Serge C. Thal
- Department of Anesthesiology, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
7
|
Reduced nasal nitric oxide levels in patients with eosinophilic chronic rhinosinusitis. Allergol Int 2019; 68:225-232. [PMID: 30348485 DOI: 10.1016/j.alit.2018.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/30/2018] [Accepted: 09/14/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In Eosinophilic chronic rhinosinusitis (ECRS), it is difficult to estimate the refractoriness and recurrence risk for each patient. Fraction of exhaled nitric oxide (FeNO) is known as a biomarker of eosinophilic inflammation in lower airway. It has been reported that nasal NO has some crucial functions in the upper and lower airways. However, in upper airway, paranasal sinuses, the usefulness of NO measurement remains controversial. The purpose of this study is to identify the usefulness of nasal NO measurement in ECRS and the involvement of nasal NO in the pathogenesis of ECRS. METHODS We compared the nasal NO levels of ECRS, non-ECRS, and normal control groups. Correlation between nasal NO levels and clinical findings were observed. Then, we compared nasal NO levels before and after endoscopic sinus surgery (ESS). We also examine whether nasal NO levels might discriminate ECRS by the receiver operating characteristic (ROC) curve analysis. RESULTS Nasal NO levels were significantly decreased in ECRS compared to the other two groups. Moreover, nasal NO levels in ECRS significantly and negatively correlated with eosinophil levels and CT score. However, they did not correlate with the nasal polyp score. Nasal NO levels were not upregulated soon after opening the sinus ostium by ESS. The ROC curves for nasal NO levels were used to discriminate all CRS patients and ECRS patients from normal controls. CONCLUSIONS Nasal NO may be useful as a marker of ECRS severity and low nasal NO levels in ECRS may contribute to its pathogenesis.
Collapse
|
8
|
Comajoan P, Gubern C, Huguet G, Serena J, Kádár E, Castellanos M. Evaluation of common housekeeping proteins under ischemic conditions and/or rt-PA treatment in bEnd.3 cells. J Proteomics 2018; 184:10-15. [PMID: 29929036 DOI: 10.1016/j.jprot.2018.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/30/2022]
Abstract
Thrombolysis with recombinant tissue plasminogen activator (rt-PA) is the only pharmacological approved treatment for ischemic stroke, despite its associated increasing risk of hemorrhagic transformation. Since many of rt-PA effects in blood-brain barrier (BBB) are not well characterized, the study of protein changes in BBB cells after rt-PA administration may help to understand its adverse effects. Our aim was to analyze protein levels of four commonly used housekeeping proteins: β-Actin, α-Tubulin, GAPDH and HPRT in bEnd.3 endothelial cell line subjected to oxygen and glucose deprivation (OGD) conditions and rt-PA treatment to determine their reliability as Western blot loading controls. bEnd.3 monolayers were subjected to 2.5 h of OGD and reperfusion with/without 20 μg/ml of rt-PA. At 3, 6, 24 and 72 h post-OGD, protein levels were analyzed by Western blot using Stain-Free technology. OGD significantly decreased β-Actin, α-Tubulin, GAPDH and HPRT protein levels at 3, 6, 24 and 72 h post-OGD without significant rt-PA treatment effects except for the GAPDH levels increase in control condition at 3 h post-OGD. The present study clearly demonstrated that β-Actin, α-Tubulin, GAPDH and HPRT proteins are not suitable as loading controls for Western Blot analysis in bEnd.3 cells after OGD. SIGNIFICANCE We reported altered levels of β-Actin, α-Tubulin, GAPDH and HPRT housekeeping proteins in bEnd.3 endothelial cell line after an ischemic insult. Therefore, we demonstrated that these proteins are not suitable as loading controls for Western Blot analysis in our experimental conditions and we recommended the use of Stain-Free gels as an alternative to traditional housekeeping proteins normalization.
Collapse
Affiliation(s)
- Pau Comajoan
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IdIBGi), Dr. Josep Trueta University Hospital, Girona, Spain; Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Girona, Spain
| | - Carme Gubern
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IdIBGi), Dr. Josep Trueta University Hospital, Girona, Spain; Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Girona, Spain
| | - Gemma Huguet
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IdIBGi), Dr. Josep Trueta University Hospital, Girona, Spain; Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Girona, Spain
| | - Joaquín Serena
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IdIBGi), Dr. Josep Trueta University Hospital, Girona, Spain; Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Girona, Spain
| | - Elisabet Kádár
- Cerebrovascular Pathology Research Group, Department of Neurology, Girona Biomedical Research Institute (IdIBGi), Dr. Josep Trueta University Hospital, Girona, Spain; Cellular and Molecular Neurobiology Research Group, Department of Biology, University of Girona (UdG), Girona, Spain.
| | - Mar Castellanos
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, A Coruña, Spain.
| |
Collapse
|
9
|
Wu J, Zhu H, Yang G, He J, Wang Y, Zhao S, Zhang X, Gui L, Zhao M, Peng S. Design and synthesis of nanoscaled IQCA-TAVV as a delivery system capable of antiplatelet activation, targeting arterial thrombus and releasing IQCA. Int J Nanomedicine 2018; 13:1139-1158. [PMID: 29520141 PMCID: PMC5833776 DOI: 10.2147/ijn.s150205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Arterial thrombosis has been associated with a series of pathological conditions, and the discovery of arterial thrombosis inhibitor is of clinical importance. METHODS By analyzing the pharmacophores of anti-platelet agents, thrombus targeting peptide and anti-thrombotic nano-systems 3S-1,2,3,4-tetrahydroisoquino-line-3-carbonyl-Thr-Ala-Arg-Gly-Asp(Val)-Val (IQCA-TAVV) was designed and prepared as a nano-scaled arterial thrombosis inhibitor. RESULTS In vitro the nanoparticles of IQCA-TAVV were able to adhere onto the surface of activated platelets, attenuate activated platelets to extend pseudopodia and inhibit activated platelets to form aggregators. In vivo IQCA-TAVV targeted arterial thrombus, dose dependently inhibited arterial thrombosis with a 1 nmol/kg of minimal effective dose, and the activity waŝ1670 folds of that of aspirin. CONCLUSION IQCA-TAVV represented the design, preparation and application of nanomedicine capable of adhering on the surface of activated platelets, attenuating platelet activation, targeting arterial thrombus and inhibiting arterial thrombosis.
Collapse
Affiliation(s)
- Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Guodong Yang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
| | - Jianhong He
- Department of Internal Medicine of TCM, The First Affiliated Hospital of Guanxi University of Chinese Medicine, Nanning, China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Liu S, Feng X, Jin R, Li G. Tissue plasminogen activator-based nanothrombolysis for ischemic stroke. Expert Opin Drug Deliv 2017; 15:173-184. [PMID: 28944694 DOI: 10.1080/17425247.2018.1384464] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Thrombolysis with intravenous tissue plasminogen activator (tPA) is the only FDA approved treatment for patients with acute ischemic stroke, but its use is limited by narrow therapeutic window, selective efficacy, and hemorrhagic complication. In the past two decades, extensive efforts have been undertaken to extend its therapeutic time window and explore alternative thrombolytic agents, but both show little progress. Nanotechnology has emerged as a promising strategy to improve the efficacy and safety of tPA. AREAS COVERED We reviewed the biology, thrombolytic mechanism, and pleiotropic functions of tPA in the brain and discussed current applications of various nanocarriers intended for the delivery of tPA for treatment of ischemic stroke. Current challenges and potential further directions of t-PA-based nanothrombolysis in stroke therapy are also discussed. EXPERT OPINION Using nanocarriers to deliver tPA offers many advantages to enhance the efficacy and safety of tPA therapy. Further research is needed to characterize the physicochemical characteristics and in vivo behavior of tPA-loaded nanocarriers. Combination of tPA based nanothrombolysis and neuroprotection represents a promising treatment strategy for acute ischemic stroke. Theranostic nanocarriers co-delivered with tPA and imaging agents are also promising for future stroke management.
Collapse
Affiliation(s)
- Shan Liu
- a Department of Neurosurgery , Pennsylvania State University College of Medicine , Hershey , PA , USA.,b Pharmaceutics Department , Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College , Beijing , PR China
| | - Xiaozhou Feng
- a Department of Neurosurgery , Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Rong Jin
- a Department of Neurosurgery , Pennsylvania State University College of Medicine , Hershey , PA , USA
| | - Guohong Li
- a Department of Neurosurgery , Pennsylvania State University College of Medicine , Hershey , PA , USA
| |
Collapse
|
11
|
Gonzalez-Gronow M, Fiedler JL, Farias Gomez C, Wang F, Ray R, Ferrell PD, Pizzo SV. Myelin basic protein stimulates plasminogen activation via tissue plasminogen activator following binding to independent l-lysine-containing domains. Biochem Biophys Res Commun 2017. [PMID: 28648598 DOI: 10.1016/j.bbrc.2017.06.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Myelin basic protein (MBP) is a key component of myelin, the specialized lipid membrane that encases the axons of all neurons. Both plasminogen (Pg) and tissue-type plasminogen activator (t-PA) bind to MBP with high affinity. We investigated the kinetics and mechanisms involved in this process using immobilized MBP and found that Pg activation by t-PA is significantly stimulated by MBP. This mechanism involves the binding of t-PA via a lysine-dependent mechanism to the Lys91 residue of the MBP NH2-terminal region Asp82 -Pro99, and the binding of Pg via a lysine-dependent mechanism to the Lys122 residue of the MBP COOH-terminal region Leu109-Gly126. In this context, MBP mimics fibrin and because MBP is a plasmin substrate, our results suggest direct participation of the Pg activation system on MBP physiology.
Collapse
Affiliation(s)
- Mario Gonzalez-Gronow
- Department of Biological Sciences, Laboratory of Environmental Neurotoxicology, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile; Department of Pathology, Duke University Medical Center, Durham, NC, USA.
| | - Jenny L Fiedler
- Laboratory of Neuroplasticity and Neurogenetics, Department of Biochemistry and Molecular Biology, Faculty of Chemistry and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Cristian Farias Gomez
- Department of Biological Sciences, Laboratory of Environmental Neurotoxicology, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, Chile
| | - Fang Wang
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Rupa Ray
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Paul D Ferrell
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Salvatore V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
12
|
Wada T, Gando S, Maekaw K, Katabami K, Sageshima H, Hayakawa M, Sawamura A. Disseminated intravascular coagulation with increased fibrinolysis during the early phase of isolated traumatic brain injury. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:219. [PMID: 28826407 PMCID: PMC5568862 DOI: 10.1186/s13054-017-1808-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/01/2017] [Indexed: 12/19/2022]
Abstract
Background There is evidence to demonstrate that the coagulopathy which occurs in patients with traumatic brain injury coincides with disseminated intravascular coagulation (DIC). We hypothesized that DIC with increased fibrinolysis during the early stage of isolated traumatic brain injury (iTBI) affects the outcome of the patients and that hypoperfusion contributes to hyperfibrinolysis in the DIC. Methods This retrospective study included 92 patients with iTBI who were divided into DIC and non-DIC groups according to the Japanese Association Acute Medicine DIC scoring system. The DIC patients were subdivided into those with and without hyperfibrinolysis. The platelet counts and global markers of coagulation and fibrinolysis were measured. Systemic inflammatory response syndrome (SIRS), organ dysfunction (assessed by the Sequential Organ Failure Assessment score), tissue hypoperfusion (assessed by the lactate levels) and the transfusion volume were also evaluated. The outcome measure was all-cause hospital mortality. Results DIC patients showed consumption coagulopathy, lower antithrombin levels and higher fibrin/fibrinogen degradation products (FDP) and D-dimer levels than non-DIC patients. All of the DIC patients developed SIRS accompanied by organ dysfunction and required higher blood transfusion volumes, leading to a worse outcome than non-DIC patients. These changes were more prominent in DIC with hyperfibrinolysis. A higher FDP/D-dimer ratio suggests that DIC belongs to the fibrinolytic phenotype and involves fibrin(ogen)olysis. The mean blood pressures of the patients with and without DIC on arrival were identical. Hypoperfusion and the lactate levels were not identified as independent predictors of hyperfibrinolysis. Conclusions DIC, especially DIC with hyperfibrinolysis, affects the outcome of patients with iTBI. Low blood pressure-induced tissue hypoperfusion does not contribute to hyperfibrinolysis in this type of DIC. Electronic supplementary material The online version of this article (doi:10.1186/s13054-017-1808-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takeshi Wada
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Satoshi Gando
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Kunihiko Maekaw
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Kenichi Katabami
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hisako Sageshima
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Mineji Hayakawa
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Atsushi Sawamura
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, N15W7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
13
|
Quattromani MJ, Pruvost M, Guerreiro C, Backlund F, Englund E, Aspberg A, Jaworski T, Hakon J, Ruscher K, Kaczmarek L, Vivien D, Wieloch T. Extracellular Matrix Modulation Is Driven by Experience-Dependent Plasticity During Stroke Recovery. Mol Neurobiol 2017; 55:2196-2213. [PMID: 28290150 PMCID: PMC5840227 DOI: 10.1007/s12035-017-0461-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/16/2017] [Indexed: 11/24/2022]
Abstract
Following stroke, complete cellular death in the ischemic brain area may ensue, with remaining brain areas undergoing tissue remodelling to various degrees. Experience-dependent brain plasticity exerted through an enriched environment (EE) promotes remodelling after central nervous system injury, such as stroke. Post-stroke tissue reorganization is modulated by growth inhibitory molecules differentially expressed within the ischemic hemisphere, like chondroitin sulfate proteoglycans found in perineuronal nets (PNNs). PNNs in the neocortex predominantly enwrap parvalbumin-containing GABAergic (PV/GABA) neurons, important in sensori-information processing. Here, we investigate how extracellular matrix (ECM) proteases and their inhibitors may participate in the regulation of PNN integrity during stroke recovery. Rats were subjected to photothrombotic stroke in the motor cortex, and functional deficits were assessed at 7 days of recovery. Sham and stroked rats were housed in either standard or EE conditions for 5 days, and infarct volumes were calculated. PNNs were visualized by immunohistochemistry and counted in the somatosensory cortex of both hemispheres. mRNA expression levels of ECM proteases and protease inhibitors were assessed by RT-qPCR and their activity analyzed by gel zymography. PNNs and protease activity were also studied in brains from stroke patients where similar results were observed. EE starting 2 days after stroke and continuing for 5 days stimulated behavioral recovery of limb-placement ability without affecting infarct size. EE promoted a decrease of PNNs around PV/GABA neurons and a concomitant modulation of the proteolytic activity and mRNA expression of ECM proteases and protease inhibitors in the somatosensory cortex. This study provides molecular targets for novel therapies that could support rehabilitation of stroke patients.
Collapse
Affiliation(s)
- Miriana Jlenia Quattromani
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden.
| | - Mathilde Pruvost
- INSERM UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, F-14074, Caen, France
| | - Carla Guerreiro
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Fredrik Backlund
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Elisabet Englund
- Division of Oncology and Pathology, Lund University Hospital, 22185, Lund, Sweden
| | - Anders Aspberg
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences, Lund University, BMC C12, 22184, Lund, Sweden
| | - Tomasz Jaworski
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Jakob Hakon
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Denis Vivien
- INSERM UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, F-14074, Caen, France.,Department of Clinical Research, Caen University Hospital, CHU Caen, 14000, Caen, France
| | - Tadeusz Wieloch
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| |
Collapse
|
14
|
Fredriksson L, Lawrence DA, Medcalf RL. tPA Modulation of the Blood-Brain Barrier: A Unifying Explanation for the Pleiotropic Effects of tPA in the CNS. Semin Thromb Hemost 2017; 43:154-168. [PMID: 27677179 PMCID: PMC5848490 DOI: 10.1055/s-0036-1586229] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The plasminogen activation (PA) system is best known for its role in fibrinolysis. However, it has also been shown to regulate many nonfibrinolytic functions in the central nervous system (CNS). In particular, tissue-type plasminogen activator (tPA) is reported to have pleiotropic activities in the CNS, regulating events such as neuronal plasticity, excitotoxicity, and cerebrovascular barrier integrity, whereas urokinase-type plasminogen activator is mainly associated with tissue remodeling and cell migration. It has been suggested that the role tPA plays in controlling barrier integrity may provide a unifying mechanism for the reported diverse, and often opposing, functions ascribed to tPA in the CNS. Here we will review the possibility that the pleiotropic effects reported for tPA in physiologic and pathologic processes in the CNS may be a consequence of its role in the neurovascular unit in regulation of cerebrovascular responses and subsequently parenchymal homeostasis. We propose that this might offer an explanation for the ongoing debate regarding the neurotoxic versus neuroprotective roles of tPA.
Collapse
Affiliation(s)
- Linda Fredriksson
- Department of Medical Biochemistry & Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Daniel A. Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| |
Collapse
|
15
|
Lemarchant S, Wojciechowski S, Vivien D, Koistinaho J. ADAMTS-4 in central nervous system pathologies. J Neurosci Res 2017; 95:1703-1711. [DOI: 10.1002/jnr.24021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 11/23/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Sighild Lemarchant
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio; University of Eastern Finland; P.O. BOX 1627 70211 Kuopio Finland
| | - Sara Wojciechowski
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio; University of Eastern Finland; P.O. BOX 1627 70211 Kuopio Finland
| | - Denis Vivien
- INSERM, INSERM UMR-S 919, “Serine Proteases and Pathophysiology of the Neurovascular Unit”; University of Caen Basse-Normandie; GIP Cyceron, Bd H. Becquerel, BP 5229 14074 Caen Cedex France
| | - Jari Koistinaho
- A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio; University of Eastern Finland; P.O. BOX 1627 70211 Kuopio Finland
| |
Collapse
|
16
|
Effect of Regulatory Element DNA Methylation on Tissue-Type Plasminogen Activator Gene Expression. PLoS One 2016; 11:e0167588. [PMID: 27973546 PMCID: PMC5156355 DOI: 10.1371/journal.pone.0167588] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/16/2016] [Indexed: 12/31/2022] Open
Abstract
Expression of the tissue-type plasminogen activator gene (t-PA; gene name PLAT) is regulated, in part, by epigenetic mechanisms. We investigated the relationship between PLAT methylation and PLAT expression in five primary human cell types and six transformed cell lines. CpG methylation was analyzed in the proximal PLAT gene promoter and near the multihormone responsive enhancer (MHRE) -7.3 kilobase pairs upstream of the PLAT transcriptional start site (TSS, -7.3 kb). In Bowes melanoma cells, the PLAT promoter and the MHRE were fully unmethylated and t-PA secretion was extremely high. In other cell types the region from -647 to -366 was fully methylated, whereas an unmethylated stretch of DNA from -121 to +94 was required but not sufficient for detectable t-PA mRNA and t-PA secretion. DNA methylation near the MHRE was not correlated with t-PA secretion. Specific methylation of the PLAT promoter region -151 to +151, inserted into a firefly luciferase reporter gene, abolished reporter gene activity. The region -121 to + 94 contains two well-described regulatory elements, a PMA-responsive element (CRE) near -106 and a GC-rich region containing an Sp1 binding site near +59. Methylation of double-stranded DNA oligonucleotides containing the CRE or the GC-rich region had little or no effect on transcription factor binding. Methylated CpGs may attract co-repressor complexes that contain histone deacetylases (HDAC). However, reporter gene activity of methylated plasmids was not restored by the HDAC inhibitor trichostatin. In conclusion, efficient PLAT gene expression requires a short stretch of unmethylated CpG sites in the proximal promoter.
Collapse
|
17
|
Lewandowski SA, Fredriksson L, Lawrence DA, Eriksson U. Pharmacological targeting of the PDGF-CC signaling pathway for blood-brain barrier restoration in neurological disorders. Pharmacol Ther 2016; 167:108-119. [PMID: 27524729 PMCID: PMC5341142 DOI: 10.1016/j.pharmthera.2016.07.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022]
Abstract
Neurological disorders account for a majority of non-malignant disability in humans and are often associated with dysfunction of the blood-brain barrier (BBB). Recent evidence shows that despite apparent variation in the origin of neural damage, the central nervous system has a common injury response mechanism involving platelet-derived growth factor (PDGF)-CC activation in the neurovascular unit and subsequent dysfunction of BBB integrity. Inhibition of PDGF-CC signaling with imatinib in mice has been shown to prevent BBB dysfunction and have neuroprotective effects in acute damage conditions, including traumatic brain injury, seizures or stroke, as well as in neurodegenerative diseases that develop over time, including multiple sclerosis and amyotrophic lateral sclerosis. Stroke and traumatic injuries are major risk factors for age-associated neurodegenerative disorders and we speculate that restoring BBB properties through PDGF-CC inhibition might provide a common therapeutic opportunity for treatment of both acute and progressive neuropathology in humans. In this review we will summarize what is known about the role of PDGF-CC in neurovascular signaling events and the variety of seemingly different neuropathologies it is involved in. We will also discuss the pharmacological means of therapeutic interventions for anti-PDGF-CC therapy and ongoing clinical trials. In summary: inhibition of PDGF-CC signaling can be protective for immediate injury and decrease the long-term neurodegenerative consequences.
Collapse
Affiliation(s)
- Sebastian A Lewandowski
- Tissue Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Scheeles v. 2, 17177, Stockholm, Sweden.
| | - Linda Fredriksson
- Vascular Biology Groups, Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Scheeles v. 2, 17177, Stockholm, Sweden; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 Medical Science Research Building III, 1150 West Medical Center Drive, Ann Arbor, MI 48109-0644, USA
| | - Daniel A Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, 7301 Medical Science Research Building III, 1150 West Medical Center Drive, Ann Arbor, MI 48109-0644, USA
| | - Ulf Eriksson
- Tissue Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Scheeles v. 2, 17177, Stockholm, Sweden.
| |
Collapse
|
18
|
Mollereau B, Rzechorzek NM, Roussel BD, Sedru M, Van den Brink DM, Bailly-Maitre B, Palladino F, Medinas DB, Domingos PM, Hunot S, Chandran S, Birman S, Baron T, Vivien D, Duarte CB, Ryoo HD, Steller H, Urano F, Chevet E, Kroemer G, Ciechanover A, Calabrese EJ, Kaufman RJ, Hetz C. Adaptive preconditioning in neurological diseases - therapeutic insights from proteostatic perturbations. Brain Res 2016; 1648:603-616. [PMID: 26923166 PMCID: PMC5010532 DOI: 10.1016/j.brainres.2016.02.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 02/06/2023]
Abstract
In neurological disorders, both acute and chronic neural stress can disrupt cellular proteostasis, resulting in the generation of pathological protein. However in most cases, neurons adapt to these proteostatic perturbations by activating a range of cellular protective and repair responses, thus maintaining cell function. These interconnected adaptive mechanisms comprise a 'proteostasis network' and include the unfolded protein response, the ubiquitin proteasome system and autophagy. Interestingly, several recent studies have shown that these adaptive responses can be stimulated by preconditioning treatments, which confer resistance to a subsequent toxic challenge - the phenomenon known as hormesis. In this review we discuss the impact of adaptive stress responses stimulated in diverse human neuropathologies including Parkinson׳s disease, Wolfram syndrome, brain ischemia, and brain cancer. Further, we examine how these responses and the molecular pathways they recruit might be exploited for therapeutic gain. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- B Mollereau
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France.
| | - N M Rzechorzek
- Centre for Clinical Brain Sciences, Chancellor's Building, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, United Kingdom
| | - B D Roussel
- Inserm, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, 14000 Caen, France
| | - M Sedru
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - D M Van den Brink
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - B Bailly-Maitre
- INSERM U1065, C3M, Team 8 (Hepatic Complications in Obesity), Nice, France
| | - F Palladino
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - D B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Faculty of Medicine, University of Chile, Santiago, Chile
| | - P M Domingos
- ITQB-UNL, Av. da Republica, EAN, 2780-157 Oeiras, Portugal
| | - S Hunot
- Inserm, U 1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - S Chandran
- Centre for Clinical Brain Sciences, Chancellor's Building, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - S Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS UMR 8249, ESPCI ParisTech, PSL Research University, 75005 Paris, France
| | - T Baron
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Neurodegenerative Diseases Unit, 31, avenue Tony Garnier, 69364 Lyon Cedex 07, France
| | - D Vivien
- Inserm, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, 14000 Caen, France
| | - C B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Faculty of Medicine, Rua Larga, and Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - H D Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - H Steller
- Howard Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - F Urano
- Washington University School of Medicine, Department of Internal Medicine, St. Louis, MO 63110 USA
| | - E Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - G Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women׳s and Children׳s Health, Karolinska University Hospital, Stockholm, Sweden
| | - A Ciechanover
- The Polak Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 30196, Israel
| | - E J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA 01003, USA
| | - R J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | - C Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Faculty of Medicine, University of Chile, Santiago, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
19
|
Appireddy R, Zerna C, Menon BK, Goyal M. Endovascular Interventions in Acute Ischemic Stroke: Recent Evidence, Current Challenges, and Future Prospects. Curr Atheroscler Rep 2016; 18:40. [DOI: 10.1007/s11883-016-0588-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
Del Bigio MR, Di Curzio DL. Nonsurgical therapy for hydrocephalus: a comprehensive and critical review. Fluids Barriers CNS 2016; 13:3. [PMID: 26846184 PMCID: PMC4743412 DOI: 10.1186/s12987-016-0025-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/15/2016] [Indexed: 12/13/2022] Open
Abstract
Pharmacological interventions have been tested experimentally and clinically to prevent hydrocephalus and avoid the need for shunting beginning in the 1950s. Clinical trials of varied quality have not demonstrated lasting and convincing protective effects through manipulation of cerebrospinal fluid production, diuresis, blood clot fibrinolysis, or manipulation of fibrosis in the subarachnoid compartment, although there remains some promise in the latter areas. Acetazolamide bolus seems to be useful for predicting shunt response in adults with hydrocephalus. Neuroprotection in the situation of established hydrocephalus has been tested experimentally beginning more recently. Therapies designed to modify blood flow or pulsation, reduce inflammation, reduce oxidative damage, or protect neurons are so far of limited success; more experimental work is needed in these areas. As has been recommended for preclinical studies in stroke and brain trauma, stringent conditions should be met for preclinical studies in hydrocephalus.
Collapse
Affiliation(s)
- Marc R Del Bigio
- Department of Pathology, University of Manitoba; Children's Hospital Research Institute of Manitoba, Diagnostic Services Manitoba, 401 Brodie Centre, 715 McDermot Avenue, Winnipeg, MB, R3E 3P5, Canada.
| | - Domenico L Di Curzio
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
21
|
Stefanitsch C, Lawrence ALE, Olverling A, Nilsson I, Fredriksson L. tPA Deficiency in Mice Leads to Rearrangement in the Cerebrovascular Tree and Cerebroventricular Malformations. Front Cell Neurosci 2015; 9:456. [PMID: 26648843 PMCID: PMC4663266 DOI: 10.3389/fncel.2015.00456] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/09/2015] [Indexed: 12/02/2022] Open
Abstract
The serine protease tissue-type plasminogen activator (tPA) is used as a thrombolytic agent in the management of ischemic stroke, but concerns for hemorrhagic conversion greatly limits the number of patients that receive this treatment. It has been suggested that the bleeding complications associated with thrombolytic tPA may be due to unanticipated roles of tPA in the brain. Recent work has suggested tPA regulation of neurovascular barrier integrity, mediated via platelet derived growth factor (PDGF)-C/PDGF receptor-α (PDGFRα) signaling, as a possible molecular mechanism affecting the outcome of stroke. To better understand the role of tPA in neurovascular regulation we conducted a detailed analysis of the cerebrovasculature in brains from adult tPA deficient (tPA−/−) mice. Our analysis demonstrates that life-long deficiency of tPA is associated with rearrangements in the cerebrovascular tree, including a reduction in the number of vascular smooth-muscle cell covered, large diameter, vessels and a decrease in vessel-associated PDGFRα expression as compared to wild-type (WT) littermate controls. In addition, we found that ablation of tPA results in an increased number of ERG-positive endothelial cells and increased junctional localization of the tight junction protein ZO1. This is intriguing since ERG is an endothelial transcription factor implicated in regulation of vascular integrity. Based on these results, we propose that the protection of barrier properties seen utilizing these tPA−/− mice might be due, at least in part, to these cerebrovascular rearrangements. In addition, we found that tPA−/− mice displayed mild cerebral ventricular malformations, a feature previously associated with ablation of PDGF-C, thereby providing an in vivo link between tPA and PDGF signaling in central nervous system (CNS) development. Taken together, the data presented here will advance our understanding of the role of tPA within the CNS and in regulation of cerebrovascular permeability.
Collapse
Affiliation(s)
- Christina Stefanitsch
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden
| | - Anna-Lisa E Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School Ann Arbor, MI, USA
| | - Anna Olverling
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden
| | - Ingrid Nilsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden
| | - Linda Fredriksson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden ; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
22
|
Hébert M, Lesept F, Vivien D, Macrez R. The story of an exceptional serine protease, tissue-type plasminogen activator (tPA). Rev Neurol (Paris) 2015; 172:186-97. [PMID: 26626577 DOI: 10.1016/j.neurol.2015.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/08/2015] [Accepted: 10/04/2015] [Indexed: 12/17/2022]
Abstract
The only acute treatment of ischemic stroke approved by the health authorities is tissue recombinant plasminogen activator (tPA)-induced thrombolysis. Under physiological conditions, tPA, belonging to the serine protease family, is secreted by endothelial and brain cells (neurons, astrocytes, microglia, oligodendrocytes). Although revascularisation induced by tPA is beneficial during a stroke, research over the past 20 years shows that tPA can also be deleterious for the brain parenchyma. Thus, in this review of the literature, after a brief history on the discovery of tPA, we reviewed current knowledge of mechanisms by which tPA can influence brain function in physiological and pathological conditions.
Collapse
Affiliation(s)
- M Hébert
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France
| | - F Lesept
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France
| | - D Vivien
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France
| | - R Macrez
- Inserm, UMR-S U919 serine proteases and pathophysiology of the neurovascular unit, 14000 Caen, France.
| |
Collapse
|
23
|
Relevance of Post-Stroke Circulating BDNF Levels as a Prognostic Biomarker of Stroke Outcome. Impact of rt-PA Treatment. PLoS One 2015; 10:e0140668. [PMID: 26469350 PMCID: PMC4607484 DOI: 10.1371/journal.pone.0140668] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 09/29/2015] [Indexed: 01/09/2023] Open
Abstract
The recombinant form of tissue plasminogen activator (rt-PA) is the only curative treatment for ischemic stroke. Recently, t-PA has been linked to the metabolism of brain-derived neurotrophic factor (BDNF), a major neurotrophin involved in post-stroke neuroplasticity. Thus, the objective of our study was to investigate the impact of rt-PA treatment on post-stroke circulating BDNF levels in humans and in animals. Serum BDNF levels and t-PA/plasmin activity were measured at hospital admission and at up to 90 days in stroke patients receiving (n = 24) or not (n = 14) rt-PA perfusion. We investigated the relationships between serum BDNF with concurrent t-PA/plasmin activity, neurological outcomes and cardiovascular scores at admission. In parallel, serum BDNF levels and t-PA/plasmin activity were assessed before and after (1, 4 and 24h) the induction of ischemic stroke in rats. Our study revealed higher serum BDNF levels and better neurological outcome in rt-PA-treated than non-treated patients. However, serum BDNF levels did not predict stroke outcome when the whole cohort of stroke patients was analyzed. By contrast, serum BDNF levels when measured at admission and at day 90 correlated with cardiovascular scores, and those at day 1 correlated with serum t-PA/plasmin activity in the whole cohort of patients whereas no association could be found in the rt-PA-treated group. In rats devoid of cardiovascular risk, no difference in post-stroke serum BDNF levels was detected between rt-PA- and vehicle-treated animals and no correlation was found between serum BDNF levels and t-PA/plasmin activity. Overall, the data suggest that serum BDNF levels may not be useful as a prognostic biomarker of stroke outcome and that endothelial dysfunction could be a confounding factor when serum BDNF levels after stroke are used to reflect of brain BDNF levels.
Collapse
|
24
|
|
25
|
Fredriksson L, Stevenson TK, Su EJ, Ragsdale M, Moore S, Craciun S, Schielke GP, Murphy GG, Lawrence DA. Identification of a neurovascular signaling pathway regulating seizures in mice. Ann Clin Transl Neurol 2015; 2:722-38. [PMID: 26273685 PMCID: PMC4531055 DOI: 10.1002/acn3.209] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/30/2015] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE A growing body of evidence suggests that increased blood-brain barrier (BBB) permeability can contribute to the development of seizures. The protease tissue plasminogen activator (tPA) has been shown to promote BBB permeability and susceptibility to seizures. In this study, we examined the pathway regulated by tPA in seizures. METHODS An experimental model of kainate-induced seizures was used in genetically modified mice, including mice deficient in tPA (tPA (-/-) ), its inhibitor neuroserpin (Nsp (-/-) ), or both (Nsp:tPA (-/-) ), and in mice conditionally deficient in the platelet-derived growth factor receptor alpha (PDGFRα). RESULTS Compared to wild-type (WT) mice, Nsp (-/-) mice have significantly reduced latency to seizure onset and generalization; whereas tPA (-/-) mice have the opposite phenotype, as do Nsp:tPA (-/-) mice. Furthermore, interventions that maintain BBB integrity delay seizure propagation, whereas osmotic disruption of the BBB in seizure-resistant tPA (-/-) mice dramatically reduces the time to seizure onset and accelerates seizure progression. The phenotypic differences in seizure progression between WT, tPA (-/-) , and Nsp (-/-) mice are also observed in electroencephalogram recordings in vivo, but absent in ex vivo electrophysiological recordings where regulation of the BBB is no longer necessary to maintain the extracellular environment. Finally, we demonstrate that these effects on seizure progression are mediated through signaling by PDGFRα on perivascular astrocytes. INTERPRETATION Together, these data identify a specific molecular pathway involving tPA-mediated PDGFRα signaling in perivascular astrocytes that regulates seizure progression through control of the BBB. Inhibition of PDGFRα signaling and maintenance of BBB integrity might therefore offer a novel clinical approach for managing seizures.
Collapse
Affiliation(s)
- Linda Fredriksson
- Division of Vascular Biology, Department of Medical Biochemistry & Biophysics, Karolinska Institutet Stockholm, Sweden ; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Tamara K Stevenson
- Department of Molecular and Integrative Physiology, University of Michigan Medical School Ann Arbor, Michigan
| | - Enming J Su
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Margaret Ragsdale
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Shannon Moore
- Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School Ann Arbor, Michigan
| | - Stefan Craciun
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Gerald P Schielke
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, University of Michigan Medical School Ann Arbor, Michigan ; Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School Ann Arbor, Michigan
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan ; Department of Molecular and Integrative Physiology, University of Michigan Medical School Ann Arbor, Michigan
| |
Collapse
|
26
|
Markers of coagulation activation and acute kidney injury in patients after hematopoietic cell transplantation. Bone Marrow Transplant 2015; 50:715-20. [PMID: 25665045 PMCID: PMC4424169 DOI: 10.1038/bmt.2015.2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 12/04/2014] [Accepted: 12/24/2014] [Indexed: 11/08/2022]
Abstract
Acute kidney injury (AKI) is common after hematopoietic cell transplant (HCT). The etiology of AKI is unknown because biopsies are rarely performed. The pathophysiology of injury is inferred from clinical data. Thrombotic microangiopathy (TMA) is often invoked as the cause of renal injury. Patients >2 years old undergoing their first HCT at Fred Hutchinson Cancer Research Center participated in this study. We prospectively measured plasma markers of coagulation activation, (PAI-1 and tPA) and fibrinolyis (D-dimer) weekly in 149 patients during the first 100 days post transplant. Cox proportional hazards modeling was used to determine associations between these markers and AKI (doubling of baseline serum creatinine). Kruskal-Wallis test was used to determine the associations between day 100 urinary albumin to creatinine ratios and these markers. Thirty one percent of patients developed AKI. Though elevations in these markers occurred frequently, neither PAI-1 nor tPA were associated with the development of AKI. D-dimer was associated with a slightly increased risk of AKI (relative risk=1.76; P-value 0.04). None of these markers were associated with micro- or macroalbuminuria at day 100. The lack of an association with AKI suggests that endothelial injury in the form of TMA is not a common cause of AKI early after transplant.
Collapse
|
27
|
Cho KS, Lee EJ, Kwon KJ, Gonzales ELT, Kim YB, Cheong JH, Bahn GH, Lee J, Han SH, Kim YT, Shin CY. Resveratrol down-regulates a glutamate-induced tissue plasminogen activator via Erk and AMPK/mTOR pathways in rat primary cortical neurons. Food Funct 2014; 5:951-60. [PMID: 24599349 DOI: 10.1039/c3fo60397k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Resveratrol (3,5,4'-trihydroxy-trans-stilbene, RSV) is a polyphenolic compound present in a variety of plant species (including grapes) that produces a myriad of biological activities including anti-inflammatory, antioxidant and neuroprotective effects. In this study, we investigate the effects of resveratrol on the basal and glutamate-stimulated expression and activity of a tissue plasminogen activator (tPA) that plays neuromodulatory or neurotoxic roles in many different neurological situations. Under basal conditions, resveratrol decreased the tPA expression and activity without affecting the tPA mRNA level in rat primary cortical neurons. RSV induced AMPK phosphorylation and inhibited mTOR phosphorylation. Inhibition of AMPK phosphorylation using compound C prevented resveratrol-induced down-regulation of tPA activity. This suggested that AMPK/mTOR-dependent translational inhibition contributes to the down-regulation of the tPA. Under glutamate-stimulated conditions of rat primary cortical neurons, tPA activity and expression were increased along with increased tPA mRNA expression but afterward treatment of RSV inhibited the glutamate-induced increase in tPA activity and expression and tPA mRNA expression. Glutamate stimulation induced activation of Akt and MAPK pathways as well as mTOR which were inhibited by RSV. Interestingly, the Erk pathway inhibitor U0126, but neither PI3K-Akt inhibitor LY294002 nor p38 inhibitor SB203580, mimicked the inhibitory action of RSV on glutamate-induced tPA up-regulation. This suggested the essential role of Erk in the transcriptional up-regulation of tPA expression, which is targeted by RSV. Glutamate stimulation induced neuronal cell death as determined by PI staining and MTT assay. However, RSV protected the cultured rat primary cortical neurons from glutamate-induced cell death as paralleled with the changes in tPA expression. These results suggested that RSV can modulate tPA activity under basal and stimulated conditions by both translational and transcriptional mechanisms. The regulation of the tPA by RSV provides additional therapeutic targets on top of the growing number of molecular substrates of RSV's action in the brain.
Collapse
Affiliation(s)
- Kyu Suk Cho
- Department of Neuroscience and Institute of Functional Genomics, Department of Pharmacology, School of Medicine, Konkuk University, 1 Hwayang-Dong, Gwangjin-Gu, Seoul 143-701, Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Gur-Wahnon D, Mizrachi T, Wald-Altman S, Higazi AAR, Brenner T. Tissue plasminogen activator involvement in experimental autoimmune myasthenia gravis: Aggravation and therapeutic potential. J Autoimmun 2014; 52:36-43. [DOI: 10.1016/j.jaut.2013.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 12/12/2013] [Indexed: 02/01/2023]
|
30
|
Lemarchant S, Pruvost M, Hébert M, Gauberti M, Hommet Y, Briens A, Maubert E, Gueye Y, Féron F, Petite D, Mersel M, do Rego JC, Vaudry H, Koistinaho J, Ali C, Agin V, Emery E, Vivien D. tPA promotes ADAMTS-4-induced CSPG degradation, thereby enhancing neuroplasticity following spinal cord injury. Neurobiol Dis 2014; 66:28-42. [PMID: 24576594 DOI: 10.1016/j.nbd.2014.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 01/23/2014] [Accepted: 02/13/2014] [Indexed: 01/12/2023] Open
Abstract
Although tissue plasminogen activator (tPA) is known to promote neuronal remodeling in the CNS, no mechanism of how this plastic function takes place has been reported so far. We provide here in vitro and in vivo demonstrations that this serine protease neutralizes inhibitory chondroitin sulfate proteoglycans (CSPGs) by promoting their degradation via the direct activation of endogenous type 4 disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS-4). Accordingly, in a model of compression-induced spinal cord injury (SCI) in rats, we found that administration of either tPA or its downstream effector ADAMTS-4 restores the tPA-dependent activity lost after the SCI and thereby, reduces content of CSPGs in the spinal cord, a cascade of events leading to an improved axonal regeneration/sprouting and eventually long term functional recovery. This is the first study to reveal a tPA-ADAMTS-4 axis and its function in the CNS. It also raises the prospect of exploiting such cooperation as a therapeutic tool for enhancing recovery after acute CNS injuries.
Collapse
Affiliation(s)
- Sighild Lemarchant
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Mathilde Pruvost
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Marie Hébert
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Maxime Gauberti
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Yannick Hommet
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Aurélien Briens
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Eric Maubert
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Yatma Gueye
- CNRS UMR-6184, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, IFR Jean Roche, Faculté de Médecine, University of Aix-Marseille, F-13916 Marseille, France
| | - François Féron
- CNRS UMR-6184, Neurobiologie des Interactions Cellulaires et Neurophysiopathologie, IFR Jean Roche, Faculté de Médecine, University of Aix-Marseille, F-13916 Marseille, France
| | - Didier Petite
- Inserm UMR-S 583, Institute for Neurosciences of Montpellier, Pathophysiology and Therapy of Sensory and Motor Deficits, Saint Eloi Hospital, F-34091 Montpellier, France
| | - Marcel Mersel
- Inserm UMR-S 583, Institute for Neurosciences of Montpellier, Pathophysiology and Therapy of Sensory and Motor Deficits, Saint Eloi Hospital, F-34091 Montpellier, France
| | - Jean-Claude do Rego
- Inserm UMR-S 982, Différenciation et Communication Neuronale et Neuroendocrine, PRIMACEN, IFRMP 23, University of Rouen, F-76130 Mont-Saint-Aignan, France
| | - Hubert Vaudry
- Inserm UMR-S 982, Différenciation et Communication Neuronale et Neuroendocrine, PRIMACEN, IFRMP 23, University of Rouen, F-76130 Mont-Saint-Aignan, France
| | - Jari Koistinaho
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, Biocenter Kuopio, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Carine Ali
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Véronique Agin
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France
| | - Evelyne Emery
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France; Department of Neurosurgery, Caen University Hospital, Avenue de la Côte de Nacre, F-14000 Caen, France.
| | - Denis Vivien
- Inserm UMR-S 919, Serine Proteases and Pathophysiology of the Neurovascular Unit, University of Caen Basse-Normandie, GIP CYCERON, F-14074 Caen Cedex, France.
| |
Collapse
|
31
|
The role of TPA I/D and PAI-1 4G/5G polymorphisms in multiple sclerosis. DISEASE MARKERS 2014; 2014:362708. [PMID: 24825926 PMCID: PMC4009184 DOI: 10.1155/2014/362708] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 03/31/2014] [Indexed: 01/05/2023]
Abstract
Background. Previous studies have shown impaired fibrinolysis in multiple sclerosis (MS) and implicated extracellular proteolytic enzymes as important factors in demyelinating neuroinflammatory disorders. Tissue-type plasminogen activator (t-PA) and its inhibitor (PAI-1) are key molecules in both fibrinolysis and extracellular proteolysis. In the present study, an association of the TPA Alu I/D and PAI-1 4G/5G polymorphisms with MS was analyzed within the Genomic Network for Multiple Sclerosis (GENoMS). Methods. The GENoMS includes four populations (Croatian, Slovenian, Serbian, and Bosnian and Herzegovinian) sharing the same geographic location and a similar ethnic background. A total of 885 patients and 656 ethnically matched healthy blood donors with no history of MS in their families were genotyped using PCR-RFLP. Results. TPA DD homozygosity was protective (OR = 0.79, 95% CI 0.63–0.99, P = 0.037) and PAI 5G5G was a risk factor for MS (OR = 1.30, 95% CI 1.01–1.66, P = 0.038). A significant effect of the genotype/carrier combination was detected in 5G5G/I carriers (OR = 1.39 95% CI 1.06–1.82, P = 0.017). Conclusions. We found a significantly harmful effect of the combination of the PAI-1 5G/5G genotype and TPA I allele on MS susceptibility, which indicates the importance of gene-gene interactions in complex diseases such as MS.
Collapse
|
32
|
Kruithof EKO, Dunoyer-Geindre S. Human tissue-type plasminogen activator. Thromb Haemost 2014; 112:243-54. [PMID: 24718307 DOI: 10.1160/th13-06-0517] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 03/07/2014] [Indexed: 11/05/2022]
Abstract
Tissue-type plasminogen activator (t-PA ) plays an important role in the removal of intravascular fibrin deposits and has several physiological roles and pathological activities in the brain. Its production by many other cell types suggests that t-PA has additional functions outside the vascular and central nervous system. Activity of t-PA is regulated at the level of its gene transcription, its mRNA stability and translation, its storage and regulated release, its interaction with cofactors that enhance its activity, its inhibition by inhibitors such as plasminogen activator inhibitor type 1 or neuroserpin, and its removal by clearance receptors. Gene transcription of t-PA is modulated by a large number of hormones, growth factors, cytokines or drugs and t-PA gene responses may be tissue-specific. The aim of this review is to summarise current knowledge on t-PA function and regulation of its pericellular activity, with an emphasis on regulation of its gene expression.
Collapse
Affiliation(s)
- E K O Kruithof
- Egbert K.O. Kruithof, Division of Angiology and Hemostasis, Department of internal medicine, University Hospital of Geneva and Faculty of Medicine of the University of Geneva, University Medical Center CMU 9094, 1 Rue Michel Servet, CH1211 Geneva 4, Switzerland, Tel.: +41 22 3795493 or +41 22 3795567, E-mail:
| | - S Dunoyer-Geindre
- Sylvie Dunoyer-Geindre, Division of Angiology and Hemostasis, Department of internal medicine, University Hospital of Geneva and Faculty of Medicine of the University of Geneva, University Medical Center CMU 9094, 1 Rue Michel Servet, CH1211 Geneva 4, Switzerland, Tel.: +41 22 3795493 or +41 22 3795567, E-mail:
| |
Collapse
|
33
|
Wang J, Zhang X, Mu L, Zhang M, Gao Z, Zhang J, Yao X, Liu C, Wang G, Wang D, Kong Q, Liu Y, Li N, Sun B, Li H. t-PA acts as a cytokine to regulate lymphocyte-endothelium adhesion in experimental autoimmune encephalomyelitis. Clin Immunol 2014; 152:90-100. [PMID: 24650778 DOI: 10.1016/j.clim.2014.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 03/10/2014] [Accepted: 03/11/2014] [Indexed: 01/17/2023]
Abstract
In this study, the capacity for t-PA to affect T cell-brain microvascular endothelial cell adhesion by acting as a cytokine was investigated. Following the treatment of a brain-derived endothelial cell line, bEnd.3, with various concentrations of t-PA, adhesion and transwell migration assays were performed. In the presence of t-PA, enhanced adhesion of T cells to bEnd.3 cells was observed. Using western blot analysis, an increase in ICAM-1 expression was detected for both t-PA-treated bEnd.3 cells and bEnd.3 cells treated with a non-enzymatic form of t-PA. In contrast, when LRP1 was blocked using a specific antibody, upregulation of ICAM-1 was inhibited and cAMP-PKA signaling was affected. Furthermore, using an EAE mouse model, administration of t-PA was associated with an increase in ICAM-1 expression by brain endothelial cells. Taken together, these findings suggest that t-PA can induce ICAM-1 expression in brain microvascular endothelial cells, and this may promote the development of EAE.
Collapse
Affiliation(s)
- Jinghua Wang
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Xin Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, China
| | - Lili Mu
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Mingqing Zhang
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Zhongming Gao
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Jia Zhang
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Xiuhua Yao
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Chuanliang Liu
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Guangyou Wang
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Dandan Wang
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Qingfei Kong
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Yumei Liu
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Na Li
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China
| | - Bo Sun
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China.
| | - Hulun Li
- Department of Neurobiology, Neurobiology Key Laboratory, Harbin Medical University, Education Department of Heilongjiang Province, Harbin, Heilongjiang 150086, China; Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
34
|
Ko HM, Joo SH, Kim P, Park JH, Kim HJ, Bahn GH, Kim HY, Lee J, Han SH, Shin CY, Park SH. Effects of Korean Red Ginseng extract on tissue plasminogen activator and plasminogen activator inhibitor-1 expression in cultured rat primary astrocytes. J Ginseng Res 2013; 37:401-12. [PMID: 24235858 PMCID: PMC3825855 DOI: 10.5142/jgr.2013.37.401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/20/2013] [Accepted: 05/24/2013] [Indexed: 01/12/2023] Open
Abstract
Korean Red Ginseng (KRG) is an oriental herbal preparation obtained from Panax ginseng Meyer (Araliaceae). To expand our understanding of the action of KRG on central nervous system (CNS) function, we examined the effects of KRG on tissue plasminogen activator (tPA)/plasminogen activator inhibitor-1 (PAI-1) expression in rat primary astrocytes. KRG extract was treated in cultured rat primary astrocytes and neuron in a concentration range of 0.1 to 1.0 mg/mL and the expression of functional tPA/PAI-1 was examined by casein zymography, Western blot and reverse transcription-polymerase chain reaction. KRG extracts increased PAI-1 expression in rat primary astrocytes in a concentration dependent manner (0.1 to 1.0 mg/mL) without affecting the expression of tPA itself. Treatment of 1.0 mg/mL KRG increased PAI-1 protein expression in rat primary astrocytes to 319.3±65.9% as compared with control. The increased PAI-1 expression mediated the overall decrease in tPA activity in rat primary astrocytes. Due to the lack of PAI-1 expression in neuron, KRG did not affect tPA activity in neuron. KRG treatment induced a concentration dependent activation of PI3K, p38, ERK1/2, and JNK in rat primary astrocytes and treatment of PI3K or MAPK inhibitors such as LY294002, U0126, SB203580, and SP600125 (10 μM each), significantly inhibited 1.0 mg/mL KRG-induced expression of PAI- 1 and down-regulation of tPA activity in rat primary astrocytes. Furthermore, compound K but not other ginsenosides such as Rb1 and Rg1 induced PAI-1 expression. KRG-induced up-regulation of PAI-1 in astrocytes may play important role in the regulation of overall tPA activity in brain, which might underlie some of the beneficial effects of KRG on CNS such as neuroprotection in ischemia and brain damaging condition as well as prevention or recovery from addiction.
Collapse
Affiliation(s)
- Hyun Myung Ko
- Department of Neuroscience, School of Medicine and Neuroscience Research Center, Institute SMART-IABS, Konkuk University, Seoul 143-701, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Aĝirbaşli MA, Song J, Lei F, Wang S, Kunselman AR, Clark JB, Myers JL, Ündar A. Comparative Effects of Pulsatile and Nonpulsatile Flow on Plasma Fibrinolytic Balance in Pediatric Patients Undergoing Cardiopulmonary Bypass. Artif Organs 2013; 38:28-33. [DOI: 10.1111/aor.12182] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Jianxun Song
- Department of Microbiology and Immunology; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
| | - Fengyang Lei
- Department of Microbiology and Immunology; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
| | - Shigang Wang
- Pediatric Cardiovascular Research Center; Penn State Hershey Children's Hospital; Penn State Milton S. Hershey Medical Center; Department of Pediatrics; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
| | - Allen R. Kunselman
- Department of Public Health Sciences; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
| | - Joseph B. Clark
- Pediatric Cardiovascular Research Center; Penn State Hershey Children's Hospital; Penn State Milton S. Hershey Medical Center; Department of Pediatrics; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
- Department of Surgery; Penn State Milton S. Hershey Medical Center; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
| | - John L. Myers
- Pediatric Cardiovascular Research Center; Penn State Hershey Children's Hospital; Penn State Milton S. Hershey Medical Center; Department of Pediatrics; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
- Department of Surgery; Penn State Milton S. Hershey Medical Center; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
| | - Akif Ündar
- Pediatric Cardiovascular Research Center; Penn State Hershey Children's Hospital; Penn State Milton S. Hershey Medical Center; Department of Pediatrics; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
- Department of Surgery; Penn State Milton S. Hershey Medical Center; Penn State Hershey College of Medicine; Pennsylvania State University; Hershey USA
- Department of Bioengineering; College of Engineering; Pennsylvania State University; University Park PA USA
| |
Collapse
|
36
|
Lavrentiadou SN, Tsantarliotou MP, Zervos IA, Nikolaidis E, Georgiadis MP, Taitzoglou IA. CCl4 induces tissue-type plasminogen activator in rat brain; protective effects of oregano, rosemary or vitamin E. Food Chem Toxicol 2013; 61:196-202. [DOI: 10.1016/j.fct.2013.06.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 06/13/2013] [Accepted: 06/24/2013] [Indexed: 01/29/2023]
|
37
|
Jullienne A, Badaut J. Molecular contributions to neurovascular unit dysfunctions after brain injuries: lessons for target-specific drug development. FUTURE NEUROLOGY 2013; 8:677-689. [PMID: 24489483 PMCID: PMC3904383 DOI: 10.2217/fnl.13.55] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The revised 'expanded' neurovascular unit (eNVU) is a physiological and functional unit encompassing endothelial cells, pericytes, smooth muscle cells, astrocytes and neurons. Ischemic stroke and traumatic brain injury are acute brain injuries directly affecting the eNVU with secondary damage, such as blood-brain barrier (BBB) disruption, edema formation and hypoperfusion. BBB dysfunctions are observed at an early postinjury time point, and are associated with eNVU activation of proteases, such as tissue plasminogen activator and matrix metalloproteinases. BBB opening is accompanied by edema formation using astrocytic AQP4 as a key protein regulating water movement. Finally, nitric oxide dysfunction plays a dual role in association with BBB injury and dysregulation of cerebral blood flow. These mechanisms are discussed including all targets of eNVU encompassing endothelium, glial cells and neurons, as well as larger blood vessels with smooth muscle. In fact, the feeding blood vessels should also be considered to treat stroke and traumatic brain injury. This review underlines the importance of the eNVU in drug development aimed at improving clinical outcome after stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jérôme Badaut
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Department of Physiology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| |
Collapse
|
38
|
Takabayashi T, Kato A, Peters AT, Hulse KE, Suh LA, Carter R, Norton J, Grammer LC, Cho SH, Tan BK, Chandra RK, Conley DB, Kern RC, Fujieda S, Schleimer RP. Excessive fibrin deposition in nasal polyps caused by fibrinolytic impairment through reduction of tissue plasminogen activator expression. Am J Respir Crit Care Med 2012; 187:49-57. [PMID: 23155140 DOI: 10.1164/rccm.201207-1292oc] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Nasal polyps (NPs) are characterized by intense edema or formation of pseudocysts filled with plasma proteins, mainly albumin. However, the mechanisms underlying NP retention of plasma proteins in their submucosa remain unclear. OBJECTIVES We hypothesized that formation of a fibrin mesh retains plasma proteins in NPs. We assessed the fibrin deposition and expression of the components of the fibrinolytic system in patients with chronic rhinosinusitis (CRS). METHODS We assessed fibrin deposition in nasal tissue from patients with CRS and control subjects by means of immunofluorescence. Fibrinolytic components, d-dimer, and plasminogen activators were measured using ELISA, real-time PCR, and immunohistochemistry. We also performed gene expression and protein quantification analysis in cultured airway epithelial cells. MEASUREMENTS AND MAIN RESULTS Immunofluorescence data showed profound fibrin deposition in NP compared with uncinate tissue (UT) from patients with CRS and control subjects. Levels of the cross-linked fibrin cleavage product protein, d-dimer, were significantly decreased in NP compared with UT from patients with CRS and control subjects, suggesting reduced fibrinolysis (P < 0.05). Expression levels of tissue plasminogen activator (t-PA) mRNA and protein were significantly decreased in NP compared with UT from patients with CRS and control subjects (P < 0.01). Immunohistochemistry demonstrated clear reduction of t-PA in NP, primarily in the epithelium and glands. Th2 cytokine-stimulated cultured airway epithelial cells showed down-regulation of t-PA, suggesting a potential Th2 mechanism in NP. CONCLUSIONS A Th2-mediated reduction of t-PA might lead to excessive fibrin deposition in the submucosa of NP, which might contribute to the tissue remodeling and pathogenesis of CRS with nasal polyps.
Collapse
Affiliation(s)
- Tetsuji Takabayashi
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sashindranath M, Sales E, Daglas M, Freeman R, Samson AL, Cops EJ, Beckham S, Galle A, McLean C, Morganti-Kossmann C, Rosenfeld JV, Madani R, Vassalli JD, Su EJ, Lawrence DA, Medcalf RL. The tissue-type plasminogen activator-plasminogen activator inhibitor 1 complex promotes neurovascular injury in brain trauma: evidence from mice and humans. ACTA ACUST UNITED AC 2012; 135:3251-64. [PMID: 22822039 DOI: 10.1093/brain/aws178] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The neurovascular unit provides a dynamic interface between the circulation and central nervous system. Disruption of neurovascular integrity occurs in numerous brain pathologies including neurotrauma and ischaemic stroke. Tissue plasminogen activator is a serine protease that converts plasminogen to plasmin, a protease that dissolves blood clots. Besides its role in fibrinolysis, tissue plasminogen activator is abundantly expressed in the brain where it mediates extracellular proteolysis. However, proteolytically active tissue plasminogen activator also promotes neurovascular disruption after ischaemic stroke; the molecular mechanisms of this process are still unclear. Tissue plasminogen activator is naturally inhibited by serine protease inhibitors (serpins): plasminogen activator inhibitor-1, neuroserpin or protease nexin-1 that results in the formation of serpin:protease complexes. Proteases and serpin:protease complexes are cleared through high-affinity binding to low-density lipoprotein receptors, but their binding to these receptors can also transmit extracellular signals across the plasma membrane. The matrix metalloproteinases are the second major proteolytic system in the mammalian brain, and like tissue plasminogen activators are pivotal to neurological function but can also degrade structures of the neurovascular unit after injury. Herein, we show that tissue plasminogen activator potentiates neurovascular damage in a dose-dependent manner in a mouse model of neurotrauma. Surprisingly, inhibition of activity following administration of plasminogen activator inhibitor-1 significantly increased cerebrovascular permeability. This led to our finding that formation of complexes between tissue plasminogen activator and plasminogen activator inhibitor-1 in the brain parenchyma facilitates post-traumatic cerebrovascular damage. We demonstrate that following trauma, the complex binds to low-density lipoprotein receptors, triggering the induction of matrix metalloproteinase-3. Accordingly, pharmacological inhibition of matrix metalloproteinase-3 attenuates neurovascular permeability and improves neurological function in injured mice. Our results are clinically relevant, because concentrations of tissue plasminogen activator: plasminogen activator inhibitor-1 complex and matrix metalloproteinase-3 are significantly elevated in cerebrospinal fluid of trauma patients and correlate with neurological outcome. In a separate study, we found that matrix metalloproteinase-3 and albumin, a marker of cerebrovascular damage, were significantly increased in brain tissue of patients with neurotrauma. Perturbation of neurovascular homeostasis causing oedema, inflammation and cell death is an important cause of acute and long-term neurological dysfunction after trauma. A role for the tissue plasminogen activator-matrix metalloproteinase axis in promoting neurovascular disruption after neurotrauma has not been described thus far. Targeting tissue plasminogen activator: plasminogen activator inhibitor-1 complex signalling or downstream matrix metalloproteinase-3 induction may provide viable therapeutic strategies to reduce cerebrovascular permeability after neurotrauma.
Collapse
Affiliation(s)
- Maithili Sashindranath
- Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3004, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|