1
|
She R, Ge J, Mei Z. New Wine in an Old Bottle: tPA for Ischemic Stroke Management. Transl Stroke Res 2025; 16:568-572. [PMID: 37921975 DOI: 10.1007/s12975-023-01209-6] [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: 10/12/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2023]
Abstract
As the only clinical thrombolytic drug approved by the FDA, tissue-type plasminogen activator (tPA) is the good standard acute treatment against ischemic stroke (IS) during the super-early stage. tPA forms the active principle of alteplase, a recombinant tissue-type plasminogen activator (rtPA), which is well known for its intravascular thrombolytic activity. However, the multifaceted functions of tPA in the central nervous system (CNS) hold untapped potential. Currently, increasing studies have explored the neuroprotective function of tPA in neurological diseases, particularly in acute ischemic stroke (AIS). A series of studies have indicated that tPA has anti-excitotoxic, neurotrophic, and anti-apoptotic effects on neurons; it is also involved in neuronal plasticity, axonal regeneration, and cerebral inflammatory processes, but how to deeply understand the underlying mechanism and take maximum advantage of tPA seems to be urgent. Therefore, more work is needed to illuminate how tPA performs with more diverse functions after stroke onset. In this comment, we focus on possible hypotheses about why and how tPA promotes ischemic neuronal survival in a comprehensive view. The text provides a holistic picture of the functions of tPA and enlists the considerations for the future, which might attract more attention toward the therapeutic potential of tPA in AIS.
Collapse
Affiliation(s)
- Ruining She
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Hunan Academy of Traditional Chinese Medicine, Changsha, Hunan, China.
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine On Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
- Third-Grade Pharmacological Laboratory On Chinese Medicine Approved By State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei, China.
| |
Collapse
|
2
|
Prunotto P, Marie P, Lebouvier L, Hommet Y, Vivien D, Ali C. The Janus face of endogenous neuronal tPA: promoting self-protection and worsening the death of neighboring neurons. Cell Death Dis 2024; 15:261. [PMID: 38609369 PMCID: PMC11014960 DOI: 10.1038/s41419-024-06655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
Recombinant tissue-type plasminogen activator (r-tPA/Actilyse) stands as the prevailing pharmacological solution for treating ischemic stroke patients, of whom because their endogenous circulating tPA alone is not sufficient to rescue reperfusion and to promote favorable outcome. Beyond the tPA contributed by circulating endothelial cells and hepatocytes, neurons also express tPA, sparking debates regarding its impact on neuronal fate ranging from pro-survival to neurotoxic properties. In order to investigate the role of neuronal tPA during brain injuries, we developed models leading to its conditional deletion in neurons, employing AAV9-pPlat-GFP and AAV9-pPlat-Cre-GFP along with tPA floxed mice. These models were subjected to N-methyl-D-aspartate (NMDA)-induced excitotoxicity or thromboembolic ischemic stroke in mice. Initially, we established that our AAV9 constructs selectively transduce neurons, bypassing other brain cell types. Subsequently, we demonstrated that tPA-expressing neurons exhibit greater resistance against NMDA-induced excitotoxicity compared to tPA negative neurons. The targeted removal of tPA in neurons heightened the susceptibility of these neurons to cell death and prevented a paracrine neurotoxic effect on tPA non-expressing neurons. Under ischemic conditions, the self-neuroprotective influence of tPA encompassed both excitatory (GFP+/Tbr1+) and inhibitory (GFP+/GABA+) neurons. Our data indicate that endogenous neuronal tPA is a protective or deleterious factor against neuronal death in an excitotoxic/ischemic context, depending on whether it acts as an autocrine or a paracrine mediator.
Collapse
Affiliation(s)
- Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Pauline Marie
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France
- Department of clinical research, CHU de Caen Normandie, Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen-Normandie, Cyceron, Caen, 14000, France.
| |
Collapse
|
3
|
Babenko VA, Fedulova KS, Silachev DN, Rahimi-Moghaddam P, Kalyuzhnaya YN, Demyanenko SV, Plotnikov EY. The Role of Matrix Metalloproteinases in Hemorrhagic Transformation in the Treatment of Stroke with Tissue Plasminogen Activator. J Pers Med 2023; 13:1175. [PMID: 37511788 PMCID: PMC10381732 DOI: 10.3390/jpm13071175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide. The only approved treatment for ischemic stroke is thrombolytic therapy with tissue plasminogen activator (tPA), though this approach often leads to a severe complication: hemorrhagic transformation (HT). The pathophysiology of HT in response to tPA is complex and not fully understood. However, numerous scientific findings suggest that the enzymatic activity and expression of matrix metalloproteinases (MMPs) in brain tissue play a crucial role. In this review article, we summarize the current knowledge of the functioning of various MMPs at different stages of ischemic stroke development and their association with HT. We also discuss the mechanisms that underlie the effect of tPA on MMPs as the main cause of the adverse effects of thrombolytic therapy. Finally, we describe recent research that aimed to develop new strategies to modulate MMP activity to improve the efficacy of thrombolytic therapy. The ultimate goal is to provide more targeted and personalized treatment options for patients with ischemic stroke to minimize complications and improve clinical outcomes.
Collapse
Affiliation(s)
- Valentina A Babenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Ksenia S Fedulova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Denis N Silachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Parvaneh Rahimi-Moghaddam
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Yulia N Kalyuzhnaya
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| | - Svetlana V Demyanenko
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| | - Egor Y Plotnikov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
4
|
Faissner A. Low-density lipoprotein receptor-related protein-1 (LRP1) in the glial lineage modulates neuronal excitability. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1190240. [PMID: 37383546 PMCID: PMC10293750 DOI: 10.3389/fnetp.2023.1190240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
The low-density lipoprotein related protein receptor 1 (LRP1), also known as CD91 or α-Macroglobulin-receptor, is a transmembrane receptor that interacts with more than 40 known ligands. It plays an important biological role as receptor of morphogens, extracellular matrix molecules, cytokines, proteases, protease inhibitors and pathogens. In the CNS, it has primarily been studied as a receptor and clearance agent of pathogenic factors such as Aβ-peptide and, lately, Tau protein that is relevant for tissue homeostasis and protection against neurodegenerative processes. Recently, it was found that LRP1 expresses the Lewis-X (Lex) carbohydrate motif and is expressed in the neural stem cell compartment. The removal of Lrp1 from the cortical radial glia compartment generates a strong phenotype with severe motor deficits, seizures and a reduced life span. The present review discusses approaches that have been taken to address the neurodevelopmental significance of LRP1 by creating novel, lineage-specific constitutive or conditional knockout mouse lines. Deficits in the stem cell compartment may be at the root of severe CNS pathologies.
Collapse
|
5
|
Beker MC, Aydinli FI, Caglayan AB, Beker M, Baygul O, Caglayan A, Popa-Wagner A, Doeppner TR, Hermann DM, Kilic E. Age-Associated Resilience Against Ischemic Injury in Mice Exposed to Transient Middle Cerebral Artery Occlusion. Mol Neurobiol 2023:10.1007/s12035-023-03353-4. [PMID: 37093494 DOI: 10.1007/s12035-023-03353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/13/2023] [Indexed: 04/25/2023]
Abstract
Ischemic stroke is the leading cause of death and disability. Although stroke mainly affects aged individuals, animal research is mostly one on young rodents. Here, we examined the development of ischemic injury in young (9-12-week-old) and adult (72-week-old) C57BL/6 and BALB/c mice exposed to 30 min of intraluminal middle cerebral artery occlusion (MCAo). Post-ischemic reperfusion did not differ between young and adult mice. Ischemic injury assessed by infarct area and blood-brain barrier (BBB) integrity assessed by IgG extravasation analysis was smaller in adult compared with young mice. Microvascular viability and neuronal survival assessed by CD31 and NeuN immunohistochemistry were higher in adult than young mice. Tissue protection was associated with stronger activation of cell survival pathways in adult than young mice. Microglial/macrophage accumulation and activation assessed by F4/80 immunohistochemistry were more restricted in adult than young mice, and pro- and anti-inflammatory cytokine and chemokine responses were reduced by aging. By means of liquid chromatography-mass spectrometry, we identified a hitherto unknown proteome profile comprising the upregulation of glycogen degradation-related pathways and the downregulation of mitochondrial dysfunction-related pathways, which distinguished post-ischemic responses of the aged compared with the young brain. Our study suggests that aging increases the brain's resilience against ischemic injury.
Collapse
Affiliation(s)
- Mustafa C Beker
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Fatmagul I Aydinli
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Medical Biology, School of Medicine, Nisantasi University, Istanbul, Turkey
| | - Ahmet B Caglayan
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Merve Beker
- Department of Medical Biology, International School of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Oguzhan Baygul
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Aysun Caglayan
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging, ARES, University of Medicine and Pharmacy Craiova, Craiova, Romania
| | | | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ertugrul Kilic
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey.
- Department of Physiology, Faculty of Medicine, Istanbul Medeniyet University, Unalan, TR-34700, Istanbul, Turkey.
| |
Collapse
|
6
|
White Matter Injury: An Emerging Potential Target for Treatment after Subarachnoid Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3842493. [PMID: 36798684 PMCID: PMC9928519 DOI: 10.1155/2023/3842493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 02/10/2023]
Abstract
Subarachnoid hemorrhage (SAH) refers to vascular brain injury mainly from a ruptured aneurysm, which has a high lifetime risk and imposes a substantial burden on patients, families, and society. Previous studies on SAH mainly focused on neurons in gray matter (GM). However, according to literature reports in recent years, in-depth research on the mechanism of white matter (WM) is of great significance to injury and recovery after SAH. In terms of functional recovery after SAH, all kinds of cells in the central nervous system (CNS) should be protected. In other words, it is necessary to protect not only GM but also WM, not only neurons but also glial cells and axons, and not only for the lesion itself but also for the prevention and treatment of remote damage. Clarifying the mechanism of white matter injury (WMI) and repair after SAH is of great importance. Therefore, this present review systematically summarizes the current research on WMI after SAH, which might provide therapeutic targets for treatment after SAH.
Collapse
|
7
|
Varangot A, Lebatard S, Bellemain-Sagnard M, Lebouvier L, Hommet Y, Vivien D. Modulations of the neuronal trafficking of tissue-type plasminogen activator (tPA) influences glutamate release. Cell Death Dis 2023; 14:34. [PMID: 36650132 PMCID: PMC9845363 DOI: 10.1038/s41419-022-05543-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
The discovery of the neuronal expression of the serine protease tissue-type plasminogen activator (tPA) has opened new avenues of research, with important implications in the physiopathology of the central nervous system. For example, the interaction of tPA with synaptic receptors (NMDAR, LRP1, Annexin II, and EGFR) and its role in the maturation of BDNF have been reported to influence synaptic plasticity and neuronal survival. However, the mechanisms regulating the neuronal trafficking of tPA are unknown. Here, using high-resolution live cell imaging and a panel of innovative genetic approaches, we first unmasked the dynamic characteristics of the dendritic and axonal trafficking of tPA-containing vesicles under different paradigms of neuronal activation or inhibition. We then report a constitutive exocytosis of tPA- and VAMP2-positive vesicles, dramatically increased in conditions of neuronal activation, with a pattern which was mainly dendritic and thus post-synaptic. We also observed that the synaptic release of tPA led to an increase of the exocytosis of VGlut1 positive vesicles containing glutamate. Finally, we described alterations of the trafficking and exocytosis of neuronal tPA in cultured cortical neurons prepared from tau-22 transgenic mice (a preclinical model of Alzheimer's disease (AD)). Altogether, these data provide new insights about the neuronal trafficking of tPA, contributing to a better knowledge of the tPA-dependent brain functions and dysfunctions.
Collapse
Affiliation(s)
- Alexandre Varangot
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Simon Lebatard
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Mathys Bellemain-Sagnard
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France.
- Department of clinical research, Caen-Normandie University Hospital, CHU, Caen, France.
| |
Collapse
|
8
|
Liang Z, Lou Y, Hao Y, Li H, Feng J, Liu S. The Relationship of Astrocytes and Microglia with Different Stages of Ischemic Stroke. Curr Neuropharmacol 2023; 21:2465-2480. [PMID: 37464832 PMCID: PMC10616922 DOI: 10.2174/1570159x21666230718104634] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 07/20/2023] Open
Abstract
Ischemic stroke is the predominant cause of severe morbidity and mortality worldwide. Post-stroke neuroinflammation has recently received increasing attention with the aim of providing a new effective treatment strategy for ischemic stroke. Microglia and astrocytes are major components of the innate immune system of the central nervous system. They can be involved in all phases of ischemic stroke, from the early stage, contributing to the first wave of neuronal cell death, to the late stage involving phagocytosis and repair. In the early stage of ischemic stroke, a vicious cycle exists between the activation of microglia and astrocytes (through astrocytic connexin 43 hemichannels), aggravating neuroinflammatory injury post-stroke. However, in the late stage of ischemic stroke, repeatedly activated microglia can induce the formation of glial scars by triggering reactive astrogliosis in the peri-infarct regions, which may limit the movement of activated microglia in reverse and restrict the diffusion of inflammation to healthy brain tissues, alleviating the neuroinflammatory injury poststroke. In this review, we elucidated the various roles of astrocytes and microglia and summarized their relationship with neuroinflammation. We also examined how astrocytes and microglia influence each other at different stages of ischemic stroke. Several potential therapeutic approaches targeting astrocytes and microglia in ischemic stroke have been reviewed. Understanding the details of astrocytemicroglia interaction processes will contribute to a better understanding of the mechanisms underlying ischemic stroke, contributing to the identification of new therapeutic interventions.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yingyue Lou
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
9
|
Seillier C, Lesept F, Toutirais O, Potzeha F, Blanc M, Vivien D. Targeting NMDA Receptors at the Neurovascular Unit: Past and Future Treatments for Central Nervous System Diseases. Int J Mol Sci 2022; 23:ijms231810336. [PMID: 36142247 PMCID: PMC9499580 DOI: 10.3390/ijms231810336] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
The excitatory neurotransmission of the central nervous system (CNS) mainly involves glutamate and its receptors, especially N-methyl-D-Aspartate receptors (NMDARs). These receptors have been extensively described on neurons and, more recently, also on other cell types. Nowadays, the study of their differential expression and function is taking a growing place in preclinical and clinical research. The diversity of NMDAR subtypes and their signaling pathways give rise to pleiotropic functions such as brain development, neuronal plasticity, maturation along with excitotoxicity, blood-brain barrier integrity, and inflammation. NMDARs have thus emerged as key targets for the treatment of neurological disorders. By their large extracellular regions and complex intracellular structures, NMDARs are modulated by a variety of endogenous and pharmacological compounds. Here, we will present an overview of NMDAR functions on neurons and other important cell types involved in the pathophysiology of neurodegenerative, neurovascular, mental, autoimmune, and neurodevelopmental diseases. We will then discuss past and future development of NMDAR targeting drugs, including innovative and promising new approaches.
Collapse
Affiliation(s)
- Célia Seillier
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
| | - Flavie Lesept
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Olivier Toutirais
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
- Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU, 14000 Caen, France
| | - Fanny Potzeha
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Manuel Blanc
- Lys Therapeutics, Cyceron, Boulevard Henri Becquerel, 14000 Caen, France
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM, GIP Cyceron, Institute Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), 14000 Caen, France
- Department of Clinical Research, Caen University Hospital, CHU, 14000 Caen, France
- Correspondence:
| |
Collapse
|
10
|
Seillier C, Hélie P, Petit G, Vivien D, Clemente D, Le Mauff B, Docagne F, Toutirais O. Roles of the tissue-type plasminogen activator in immune response. Cell Immunol 2021; 371:104451. [PMID: 34781155 PMCID: PMC8577548 DOI: 10.1016/j.cellimm.2021.104451] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/06/2021] [Accepted: 10/29/2021] [Indexed: 11/30/2022]
Abstract
The COVID-19 pandemic has once again
brought to the forefront the existence of a tight link between the
coagulation/fibrinolytic system and the immunologic processes.
Tissue-type plasminogen activator (tPA) is a serine protease with a key
role in fibrinolysis by converting plasminogen into plasmin that can
finally degrade fibrin clots. tPA is released in the blood by endothelial
cells and hepatocytes but is also produced by various types of immune
cells including T cells and monocytes. Beyond its role on hemostasis, tPA
is also a potent modulator of inflammation and is involved in the
regulation of several inflammatory diseases. Here, after a brief
description of tPA structure, we review its new functions in adaptive
immunity focusing on T cells and antigen presenting cells. We intend to
synthesize the recent knowledge on proteolysis- and receptor-mediated
effects of tPA on immune response in physiological and pathological
context.
Collapse
Affiliation(s)
- Célia Seillier
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Pauline Hélie
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Gautier Petit
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Clinical Research, Caen University Hospital, CHU Caen, France
| | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Brigitte Le Mauff
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France
| | - Fabian Docagne
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France
| | - Olivier Toutirais
- Normandie Univ, UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Caen, France; Department of Immunology and Histocompatibility (HLA), Caen University Hospital, CHU Caen, France.
| |
Collapse
|
11
|
Garnier E, Levard D, Ali C, Buendia I, Hommet Y, Gauberti M, Crepaldi T, Comoglio P, Rubio M, Vivien D, Docagne F, Martinez de Lizarrondo S. Factor XII protects neurons from apoptosis by epidermal and hepatocyte growth factor receptor-dependent mechanisms. J Thromb Haemost 2021; 19:2235-2247. [PMID: 34060720 DOI: 10.1111/jth.15414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Factor XII (FXII) is a serine protease that participates in the intrinsic coagulation pathway. Several studies have shown that plasma FXII exerts a deleterious role in cerebral ischemia and traumatic brain injury by promoting thrombo-inflammation. Nevertheless, the impact of FXII on neuronal cell fate remains unknown. OBJECTIVES We investigated the role of FXII and FXIIa in neuronal injury and apoptotic cell death. METHODS We tested the neuroprotective roles of FXII and FXIIa in an experimental model of neuronal injury induced by stereotaxic intracerebral injection of N-methyl-D-aspartic acid (NMDA) in vivo and in a model of apoptotic death of murine primary neuronal cultures through serum deprivation in vitro. RESULTS Here, we found that exogenous FXII and FXIIa reduce brain lesions induced by NMDA injection in vivo. Furthermore, FXII protects cultured neurons from apoptosis through a growth factor--like effect. This mechanism was triggered by direct interaction with epidermal growth factor (EGF) receptor and subsequent activation of this receptor. Interestingly, the "proteolytically" active and two-chain form of FXII, FXIIa, exerts its protective effects by an alternative signaling pathway. FXIIa activates the pro-form of hepatocyte growth factor (HGF) into HGF, which in turn activated the HGF receptor (HGFR) pathway. CONCLUSION This study describes two novel mechanisms of action of FXII and identifies neurons as target cells for the protective effects of single and two-chain forms of FXII. Therefore, inhibition of FXII in neurological disorders may have deleterious effects by preventing its beneficial effects on neuronal survival.
Collapse
Affiliation(s)
- Eugénie Garnier
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Damien Levard
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Carine Ali
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Izaskun Buendia
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Yannick Hommet
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Maxime Gauberti
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Tiziana Crepaldi
- Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino Medical School, Candiolo, Italy
| | | | - Marina Rubio
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Denis Vivien
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
- CHU Caen, Department of Clinical Research, CHU Caen Côte de Nacre, Caen, France
| | - Fabian Docagne
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| | - Sara Martinez de Lizarrondo
- Normandie Univ, Unicaen, Inserm, Physiopathology and imaging of neurological disorders (PhIND), Caen, France
| |
Collapse
|
12
|
Hélie P, Camacho-Toledano C, Lesec L, Seillier C, Miralles AJ, Ortega MC, Guérit S, Lebas H, Bardou I, Vila-Del Sol V, Vivien D, Le Mauff B, Clemente D, Docagne F, Toutirais O. Tissue plasminogen activator worsens experimental autoimmune encephalomyelitis by complementary actions on lymphoid and myeloid cell responses. J Neuroinflammation 2021; 18:52. [PMID: 33610187 PMCID: PMC7897384 DOI: 10.1186/s12974-021-02102-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 02/03/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Tissue plasminogen activator (tPA) is a serine protease involved in fibrinolysis. It is released by endothelial cells, but also expressed by neurons and glial cells in the central nervous system (CNS). Interestingly, this enzyme also contributes to pathological processes in the CNS such as neuroinflammation by activating microglia and increasing blood-brain barrier permeability. Nevertheless, its role in the control of adaptive and innate immune response remains poorly understood. METHODS tPA effects on myeloid and lymphoid cell response were studied in vivo in the mouse model of multiple sclerosis experimental autoimmune encephalomyelitis and in vitro in splenocytes. RESULTS tPA-/- animals exhibited less severe experimental autoimmune encephalomyelitis than their wild-type counterparts. This was accompanied by a reduction in both lymphoid and myeloid cell populations in the spinal cord parenchyma. In parallel, tPA increased T cell activation and proliferation, as well as cytokine production by a protease-dependent mechanism and via plasmin generation. In addition, tPA directly raised the expression of MHC-II and the co-stimulatory molecules CD80 and CD86 at the surface of dendritic cells and macrophages by a direct action dependent of the activation of epidermal growth factor receptor. CONCLUSIONS Our study provides new insights into the mechanisms responsible for the harmful functions of tPA in multiple sclerosis and its animal models: tPA promotes the proliferation and activation of both lymphoid and myeloid populations by distinct, though complementary, mechanisms.
Collapse
Affiliation(s)
- Pauline Hélie
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
- Present address: Theodor Kocher Institute, University of Bern, Freiestrasse 1, CH-3012, Bern, Switzerland
| | - Celia Camacho-Toledano
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071, Toledo, Spain
| | - Léonie Lesec
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
| | - Célia Seillier
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
| | - Antonio J Miralles
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071, Toledo, Spain
| | - Maria Cristina Ortega
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071, Toledo, Spain
| | - Sylvaine Guérit
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
| | - Héloïse Lebas
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
| | - Isabelle Bardou
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
| | | | - Denis Vivien
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
- Department of Clinical Research, Caen University Hospital, CHU, Caen, France
| | - Brigitte Le Mauff
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
- Department of Immunology and Immunopathology, Caen University Hospital, CHU, Caen, France
| | - Diego Clemente
- Grupo de Neuroinmuno-Reparación, Hospital Nacional de Parapléjicos, Finca La Peraleda s/n, 45071, Toledo, Spain
| | - Fabian Docagne
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France.
| | - Olivier Toutirais
- UNICAEN, INSERM, GIP Cyceron, Institut Blood and Brain @Caen-Normandie (BB@C), UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie Univ, Caen, France
- Department of Immunology and Immunopathology, Caen University Hospital, CHU, Caen, France
| |
Collapse
|
13
|
Nikitin D, Choi S, Mican J, Toul M, Ryu WS, Damborsky J, Mikulik R, Kim DE. Development and Testing of Thrombolytics in Stroke. J Stroke 2021; 23:12-36. [PMID: 33600700 PMCID: PMC7900387 DOI: 10.5853/jos.2020.03349] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Despite recent advances in recanalization therapy, mechanical thrombectomy will never be a treatment for every ischemic stroke because access to mechanical thrombectomy is still limited in many countries. Moreover, many ischemic strokes are caused by occlusion of cerebral arteries that cannot be reached by intra-arterial catheters. Reperfusion using thrombolytic agents will therefore remain an important therapy for hyperacute ischemic stroke. However, thrombolytic drugs have shown limited efficacy and notable hemorrhagic complication rates, leaving room for improvement. A comprehensive understanding of basic and clinical research pipelines as well as the current status of thrombolytic therapy will help facilitate the development of new thrombolytics. Compared with alteplase, an ideal thrombolytic agent is expected to provide faster reperfusion in more patients; prevent re-occlusions; have higher fibrin specificity for selective activation of clot-bound plasminogen to decrease bleeding complications; be retained in the blood for a longer time to minimize dosage and allow administration as a single bolus; be more resistant to inhibitors; and be less antigenic for repetitive usage. Here, we review the currently available thrombolytics, strategies for the development of new clot-dissolving substances, and the assessment of thrombolytic efficacies in vitro and in vivo.
Collapse
Affiliation(s)
- Dmitri Nikitin
- International Centre for Clinical Research, St. Anne's Hospital, Brno, Czech Republic.,Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Seungbum Choi
- Molecular Imaging and Neurovascular Research Laboratory, Department of Neurology, Dongguk University College of Medicine, Goyang, Korea
| | - Jan Mican
- International Centre for Clinical Research, St. Anne's Hospital, Brno, Czech Republic.,Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Neurology, St. Anne's Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Toul
- International Centre for Clinical Research, St. Anne's Hospital, Brno, Czech Republic.,Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Wi-Sun Ryu
- Department of Neurology, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Jiri Damborsky
- International Centre for Clinical Research, St. Anne's Hospital, Brno, Czech Republic.,Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Robert Mikulik
- International Centre for Clinical Research, St. Anne's Hospital, Brno, Czech Republic.,Department of Neurology, St. Anne's Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dong-Eog Kim
- Molecular Imaging and Neurovascular Research Laboratory, Department of Neurology, Dongguk University College of Medicine, Goyang, Korea.,Department of Neurology, Dongguk University Ilsan Hospital, Goyang, Korea
| |
Collapse
|
14
|
Pasquet N, Douceau S, Naveau M, Lesept F, Louessard M, Lebouvier L, Hommet Y, Vivien D, Bardou I. Tissue-Type Plasminogen Activator Controlled Corticogenesis Through a Mechanism Dependent of NMDA Receptors Expressed on Radial Glial Cells. Cereb Cortex 2020; 29:2482-2498. [PMID: 29878094 DOI: 10.1093/cercor/bhy119] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Indexed: 01/24/2023] Open
Abstract
Modifications of neuronal migration during development, including processes that control cortical lamination are associated with functional deficits at adult stage. Here, we report for the first time that the lack of the serine protease tissue-type Plasminogen Activator (tPA), previously characterized as a neuromodulator and a gliotransmitter, leads to an altered cortical lamination in adult. This results in a neuronal migration defect of tPA deficient neurons which are stopped in the intermediate zone at E16. This phenotype is rescued by re-expressing a wild-type tPA in cortical neurons at E14 but not by a tPA that cannot interact with NMDAR. We thus hypothetized that the tPA produced by cortical neuronal progenitors can control their own radial migration through a mechanism dependent of NMDAR expressed at the surface of radial glial cells (RGC). Accordingly, conditional deletion of tPA in neuronal progenitors at E14 or overexpression of a dominant-negative NMDAR that cannot bind tPA in RGC also delayed neuronal migration. Moreover, the lack of tPA lead to an impaired maturation and orientation of RGC. These data provide the first demonstration that the neuronal serine protease tPA is an actor of a proper corticogenesis by its ability to control NMDAR signaling in RGC.
Collapse
Affiliation(s)
- Nolwenn Pasquet
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Sara Douceau
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Mickael Naveau
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Flavie Lesept
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Morgane Louessard
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Laurent Lebouvier
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Yannick Hommet
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France.,CHU Caen, Clinical Research Department, Caen University Hospital, Caen, France
| | - Isabelle Bardou
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| |
Collapse
|
15
|
Extrinsic Factors Driving Oligodendrocyte Lineage Cell Progression in CNS Development and Injury. Neurochem Res 2020; 45:630-642. [PMID: 31997102 PMCID: PMC7058689 DOI: 10.1007/s11064-020-02967-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.
Collapse
|
16
|
Palmer JA, Wheaton LA, Gray WA, Saltão da Silva MA, Wolf SL, Borich MR. Role of Interhemispheric Cortical Interactions in Poststroke Motor Function. Neurorehabil Neural Repair 2019; 33:762-774. [PMID: 31328638 DOI: 10.1177/1545968319862552] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background/Objective. We investigated interhemispheric interactions in stroke survivors by measuring transcranial magnetic stimulation (TMS)-evoked cortical coherence. We tested the effect of TMS on interhemispheric coherence during rest and active muscle contraction and compared coherence in stroke and older adults. We evaluated the relationships between interhemispheric coherence, paretic motor function, and the ipsilateral cortical silent period (iSP). Methods. Participants with (n = 19) and without (n = 14) chronic stroke either rested or maintained a contraction of the ipsilateral hand muscle during simultaneous recordings of evoked responses to TMS of the ipsilesional/nondominant (i/ndM1) and contralesional/dominant (c/dM1) primary motor cortex with EEG and in the hand muscle with EMG. We calculated pre- and post-TMS interhemispheric beta coherence (15-30 Hz) between motor areas in both conditions and the iSP duration during the active condition. Results. During active i/ndM1 TMS, interhemispheric coherence increased immediately following TMS in controls but not in stroke. Coherence during active cM1 TMS was greater than iM1 TMS in the stroke group. Coherence during active iM1 TMS was less in stroke participants and was negatively associated with measures of paretic arm motor function. Paretic iSP was longer compared with controls and negatively associated with clinical measures of manual dexterity. There was no relationship between coherence and. iSP for either group. No within- or between-group differences in coherence were observed at rest. Conclusions. TMS-evoked cortical coherence during hand muscle activation can index interhemispheric interactions associated with poststroke motor function and potentially offer new insights into neural mechanisms influencing functional recovery.
Collapse
Affiliation(s)
| | | | | | | | - Steven L Wolf
- 1 Emory University, Atlanta, GA, USA
- 2 Atlanta VA Visual and Neurocognitive Center of Excellence, Decatur, GA, USA
| | | |
Collapse
|
17
|
Mican J, Toul M, Bednar D, Damborsky J. Structural Biology and Protein Engineering of Thrombolytics. Comput Struct Biotechnol J 2019; 17:917-938. [PMID: 31360331 PMCID: PMC6637190 DOI: 10.1016/j.csbj.2019.06.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
Myocardial infarction and ischemic stroke are the most frequent causes of death or disability worldwide. Due to their ability to dissolve blood clots, the thrombolytics are frequently used for their treatment. Improving the effectiveness of thrombolytics for clinical uses is of great interest. The knowledge of the multiple roles of the endogenous thrombolytics and the fibrinolytic system grows continuously. The effects of thrombolytics on the alteration of the nervous system and the regulation of the cell migration offer promising novel uses for treating neurodegenerative disorders or targeting cancer metastasis. However, secondary activities of thrombolytics may lead to life-threatening side-effects such as intracranial bleeding and neurotoxicity. Here we provide a structural biology perspective on various thrombolytic enzymes and their key properties: (i) effectiveness of clot lysis, (ii) affinity and specificity towards fibrin, (iii) biological half-life, (iv) mechanisms of activation/inhibition, and (v) risks of side effects. This information needs to be carefully considered while establishing protein engineering strategies aiming at the development of novel thrombolytics. Current trends and perspectives are discussed, including the screening for novel enzymes and small molecules, the enhancement of fibrin specificity by protein engineering, the suppression of interactions with native receptors, liposomal encapsulation and targeted release, the application of adjuvants, and the development of improved production systems.
Collapse
Key Words
- EGF, Epidermal growth factor domain
- F, Fibrin binding finger domain
- Fibrinolysis
- K, Kringle domain
- LRP1, Low-density lipoprotein receptor-related protein 1
- MR, Mannose receptor
- NMDAR, N-methyl-D-aspartate receptor
- P, Proteolytic domain
- PAI-1, Inhibitor of tissue plasminogen activator
- Plg, Plasminogen
- Plm, Plasmin
- RAP, Receptor antagonist protein
- SAK, Staphylokinase
- SK, Streptokinase
- Staphylokinase
- Streptokinase
- Thrombolysis
- Tissue plasminogen activator
- Urokinase
- t-PA, Tissue plasminogen activator
Collapse
Affiliation(s)
- Jan Mican
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Martin Toul
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - David Bednar
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| | - Jiri Damborsky
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Masaryk University, Kamenice 5/A13, 625 00 Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
| |
Collapse
|
18
|
The interleukin-4/PPARγ signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury. PLoS Biol 2019; 17:e3000330. [PMID: 31226122 PMCID: PMC6608986 DOI: 10.1371/journal.pbio.3000330] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/03/2019] [Accepted: 05/31/2019] [Indexed: 11/29/2022] Open
Abstract
The repair of white matter damage is of paramount importance for functional recovery after brain injuries. Here, we report that interleukin-4 (IL-4) promotes oligodendrocyte regeneration and remyelination. IL-4 receptor expression was detected in a variety of glial cells after ischemic brain injury, including oligodendrocyte lineage cells. IL-4 deficiency in knockout mice resulted in greater deterioration of white matter over 14 d after stroke. Consistent with these findings, intranasal delivery of IL-4 nanoparticles after stroke improved white matter integrity and attenuated long-term sensorimotor and cognitive deficits in wild-type mice, as revealed by histological immunostaining, electron microscopy, diffusion tensor imaging, and electrophysiology. The selective effect of IL-4 on remyelination was verified in an ex vivo organotypic model of demyelination. By leveraging primary oligodendrocyte progenitor cells (OPCs), microglia-depleted mice, and conditional OPC-specific peroxisome proliferator-activated receptor gamma (PPARγ) knockout mice, we discovered a direct salutary effect of IL-4 on oligodendrocyte differentiation that was mediated by the PPARγ axis. Our findings reveal a new regenerative role of IL-4 in the central nervous system (CNS), which lies beyond its known immunoregulatory functions on microglia/macrophages or peripheral lymphocytes. Therefore, intranasal IL-4 delivery may represent a novel therapeutic strategy to improve white matter integrity in stroke and other brain injuries. The repair and remyelination of white matter are of paramount importance for functional recovery after brain injuries. This study shows that interleukin-4 plays an essential role in oligodendrocyte differentiation and long-term white matter recovery, beyond its well-known immunoregulatory functions, and is mediated by the PPARγ axis.
Collapse
|
19
|
T-Regulatory Cells Confer Increased Myelination and Stem Cell Activity after Stroke-Induced White Matter Injury. J Clin Med 2019; 8:jcm8040537. [PMID: 31010132 PMCID: PMC6518209 DOI: 10.3390/jcm8040537] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/05/2019] [Accepted: 04/17/2019] [Indexed: 12/29/2022] Open
Abstract
Stroke-induced hypoxia causes oligodendrocyte death due to inflammation, lack of oxygen and exacerbation of cell death. Bone marrow-derived stem cells (BMSCs) possess an endogenous population of T-regulatory cells (Tregs) which reduce secretion of pro-inflammatory cytokines that lead to secondary cell death. Here, we hypothesize that oligodendrocyte progenitor cells (OPCs) cultured with BMSCs containing their native Treg population show greater cell viability, less pro-inflammatory cytokine secretion and greater myelin production after exposure to oxygen-glucose deprivation and reoxygenation (OGD/R) than OPCs cultured without Tregs. OPCs were cultured and then exposed to OGD/R. BMSCs with or without Tregs were added to the co-culture immediately after ischemia. The Tregs were depleted by running the BMSCs through a column containing a magnetic substrate. Fibroblast growth factor beta (FGF-β) and interleukin 6 (IL-6) ELISAs determined BMSC activity levels. Immunohistochemistry assessed OPC differentiation. OPCs cultured with BMSCs containing their endogenous Tregs showed increased myelin production compared to the BMSCs with depleted Tregs. IL-6 and FGF-β were increased in the group cultured with Tregs. Collectively, these results suggest that BMSCs containing Tregs are more therapeutically active, and that Tregs have beneficial effects on OPCs subjected to ischemia. Tregs play an important role in stem cell therapy and can potentially treat white matter injury post-stroke.
Collapse
|
20
|
Protease-independent action of tissue plasminogen activator in brain plasticity and neurological recovery after ischemic stroke. Proc Natl Acad Sci U S A 2019; 116:9115-9124. [PMID: 30996120 DOI: 10.1073/pnas.1821979116] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that tissue plasminogen activator (tPA), currently the only FDA-approved medication for ischemic stroke, exerts important biological actions on the CNS besides its well-known thrombolytic effect. In this study, we investigated the role of tPA on primary neurons in culture and on brain recovery and plasticity after ischemic stroke in mice. Treatment with recombinant tPA stimulated axonal growth in culture, an effect independent of its protease activity and achieved through epidermal growth factor receptor (EGFR) signaling. After permanent focal cerebral ischemia, tPA knockout mice developed more severe sensorimotor and cognitive deficits and greater axonal and myelin injury than wild-type mice, suggesting that endogenously expressed tPA promotes long-term neurological recovery after stroke. In tPA knockout mice, intranasal administration of recombinant tPA protein 6 hours poststroke and 7 more times at 2 d intervals mitigated white matter injury, improved axonal conduction, and enhanced neurological recovery. Consistent with the proaxonal growth effects observed in vitro, exogenous tPA delivery increased poststroke axonal sprouting of corticobulbar and corticospinal tracts, which might have contributed to restoration of neurological functions. Notably, recombinant mutant tPA-S478A lacking protease activity (but retaining the EGF-like domain) was as effective as wild-type tPA in rescuing neurological functions in tPA knockout stroke mice. These findings demonstrate that tPA improves long-term functional outcomes in a clinically relevant stroke model, likely by promoting brain plasticity through EGFR signaling. Therefore, treatment with the protease-dead recombinant tPA-S478A holds particular promise as a neurorestorative therapy, as the risk for triggering intracranial hemorrhage is eliminated and tPA-S478A can be delivered intranasally hours after stroke.
Collapse
|
21
|
Zhu J, Wan Y, Xu H, Wu Y, Hu B, Jin H. The role of endogenous tissue-type plasminogen activator in neuronal survival after ischemic stroke: friend or foe? Cell Mol Life Sci 2019; 76:1489-1506. [PMID: 30656378 PMCID: PMC11105644 DOI: 10.1007/s00018-019-03005-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/19/2018] [Accepted: 01/03/2019] [Indexed: 12/29/2022]
Abstract
Endogenous protease tissue-type plasminogen activator (tPA) has highly efficient fibrinolytic activity and its recombinant variants alteplase and tenecteplase are established as highly effective thrombolytic drugs for ischemic stroke. Endogenous tPA is constituted of five functional domains through which it interacts with a variety of substrates, binding proteins and receptors, thus having enzymatic and cytokine-like effects to act on all cell types of the brain. In the past 2 decades, numerous studies have explored the clinical relevance of endogenous tPA in neurological diseases, especially in ischemic stroke. tPA is released from many cells within the brain parenchyma exposed to ischemia conditions in vitro and in vivo, which is believed to control neuronal fate. Some studies proved that tPA could induce blood-brain barrier disruption, neural excitotoxicity and inflammation, while others indicated that tPA also has anti-excitotoxic, neurotrophic and anti-apoptotic effects on neurons. Therefore, more work is needed to elucidate how tPA mediates such opposing functions that may amplify tPA from a therapeutic means into a key therapeutic target in endogenous neuroprotection after stroke. In this review, we summarize the biological characteristics and pleiotropic functions of tPA in the brain. Then we focus on possible hypotheses about why and how endogenous tPA mediates ischemic neuronal death and survival. Finally, we analyze how endogenous tPA affects neuron fate in ischemic stroke in a comprehensive view.
Collapse
Affiliation(s)
- Jiayi Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yan Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Hexiang Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yulang Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| | - Huijuan Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
22
|
Bres EE, Faissner A. Low Density Receptor-Related Protein 1 Interactions With the Extracellular Matrix: More Than Meets the Eye. Front Cell Dev Biol 2019; 7:31. [PMID: 30931303 PMCID: PMC6428713 DOI: 10.3389/fcell.2019.00031] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a biological substrate composed of collagens, proteoglycans and glycoproteins that ensures proper cell migration and adhesion and keeps the cell architecture intact. The regulation of the ECM composition is a vital process strictly controlled by, among others, proteases, growth factors and adhesion receptors. As it appears, ECM remodeling is also essential for proper neuronal and glial development and the establishment of adequate synaptic signaling. Hence, disturbances in ECM functioning are often present in neurodegenerative diseases like Alzheimer’s disease. Moreover, mutations in ECM molecules are found in some forms of epilepsy and malfunctioning of ECM-related genes and pathways can be seen in, for example, cancer or ischemic injury. Low density lipoprotein receptor-related protein 1 (Lrp1) is a member of the low density lipoprotein receptor family. Lrp1 is involved not only in ligand uptake, receptor mediated endocytosis and lipoprotein transport—functions shared by low density lipoprotein receptor family members—but also regulates cell surface protease activity, controls cellular entry and binding of toxins and viruses, protects against atherosclerosis and acts on many cell signaling pathways. Given the plethora of functions, it is not surprising that Lrp1 also impacts the ECM and is involved in its remodeling. This review focuses on the role of Lrp1 and some of its major ligands on ECM function. Specifically, interactions with two Lrp1 ligands, integrins and tissue plasminogen activator are described in more detail.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
23
|
Peng X, Zhao J, Liu J, Li S. Advances in biomarkers of cerebral small vessel disease. JOURNAL OF NEURORESTORATOLOGY 2019. [DOI: 10.26599/jnr.2019.9040021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a type of syndrome caused by lesions in perforating arteries, small veins, small arteries, or capillaries, resulting in clinical, imaging, or pathological alterations. The occurrence and development of CSVD are related to various cerebrovascular risk factors, such as metabolism and genetic factors. CSVD is diagnosed based on brain imaging biomarkers; however, biomarkers capable of predicting and diagnosing CSVD early in its progression have not been found. Exploring biomarkers closely related to disease progression is of great significance for early diagnosis, prognosis, prevention, and treatment of CSVD. This article examines the research progress of CSVD biomarkers, from inflammatory biomarkers, coagulation and fibrinolysis markers, biomarkers of endothelial dysfunction, biomarkers related to cerebrospinal fluid, and gene markers.
Collapse
|
24
|
Thiebaut AM, Gauberti M, Ali C, Martinez De Lizarrondo S, Vivien D, Yepes M, Roussel BD. The role of plasminogen activators in stroke treatment: fibrinolysis and beyond. Lancet Neurol 2018; 17:1121-1132. [PMID: 30507392 DOI: 10.1016/s1474-4422(18)30323-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/25/2018] [Accepted: 08/28/2018] [Indexed: 12/20/2022]
Abstract
Although recent technical advances in thrombectomy have revolutionised acute stroke treatment, prevalence of disability and death related to stroke remain high. Therefore, plasminogen activators-eukaryotic, bacterial, or engineered forms that can promote fibrinolysis by converting plasminogen into active plasmin and facilitate clot breakdown-are still commonly used in the acute treatment of ischaemic stroke. Hence, plasminogen activators have become a crucial area for clinical investigation for their ability to recanalise occluded arteries in ischaemic stroke and to accelerate haematoma clearance in haemorrhagic stroke. However, inconsistent results, insufficient evidence of efficacy, or reports of side-effects in trial settings might reduce the use of plasminogen activators in clinical practice. Additionally, the mechanism of action for plasminogen activators could extend beyond the vessel lumen and involve plasminogen-independent processes, which would suggest that plasminogen activators have also non-fibrinolytic roles. Understanding the complex mechanisms of action of plasminogen activators can guide future directions for therapeutic interventions in patients with stroke.
Collapse
Affiliation(s)
- Audrey M Thiebaut
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Maxime Gauberti
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Carine Ali
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Sara Martinez De Lizarrondo
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France; Clinical Research Department, University Hospital Caen-Normandy, Caen, France
| | - Manuel Yepes
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, and Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| | - Benoit D Roussel
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Cyceron, Caen, France.
| |
Collapse
|
25
|
Tissue plasminogen activator promotes white matter integrity and functional recovery in a murine model of traumatic brain injury. Proc Natl Acad Sci U S A 2018; 115:E9230-E9238. [PMID: 30201709 DOI: 10.1073/pnas.1810693115] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recombinant tissue plasminogen activator (tPA) is a Food and Drug Administration-approved thrombolytic treatment for ischemic stroke. tPA is also naturally expressed in glial and neuronal cells of the brain, where it promotes axon outgrowth and synaptic plasticity. However, there are conflicting reports of harmful versus neuroprotective effects of tPA in acute brain injury models. Furthermore, its impact on white matter integrity in preclinical traumatic brain injury (TBI) has not been thoroughly explored, although white matter disruption is a better predictor of long-term clinical outcomes than focal lesion volumes. Here we show that the absence of endogenous tPA in knockout mice impedes long-term recovery of white matter and neurological function after TBI. tPA-knockout mice exhibited greater asymmetries in forepaw use, poorer sensorimotor balance and coordination, and inferior spatial learning and memory up to 35 d after TBI. White matter damage was also more prominent in tPA knockouts, as shown by diffusion tensor imaging, histological criteria, and electrophysiological assessments of axon conduction properties. Replenishment of tPA through intranasal application of the recombinant protein in tPA-knockout mice enhanced neurological function, the structural and functional integrity of white matter, and postinjury compensatory sprouting in corticofugal projections. tPA also promoted neurite outgrowth in vitro, partly through the epidermal growth factor receptor. Both endogenous and exogenous tPA protected against white matter injury after TBI without increasing intracerebral hemorrhage volumes. These results unveil a previously unappreciated role for tPA in the protection and/or repair of white matter and long-term functional recovery after TBI.
Collapse
|
26
|
Gauberti M, Potzeha F, Vivien D, Martinez de Lizarrondo S. Impact of Bradykinin Generation During Thrombolysis in Ischemic Stroke. Front Med (Lausanne) 2018; 5:195. [PMID: 30018956 PMCID: PMC6037726 DOI: 10.3389/fmed.2018.00195] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/14/2018] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. Current medical management in the acute phase is based on the activation of the fibrinolytic cascade by intravenous injection of a plasminogen activator (such as tissue-type plasminogen activator, tPA) that promotes restauration of the cerebral blood flow and improves stroke outcome. Unfortunately, the use of tPA is associated with deleterious effects such as hemorrhagic transformation, symptomatic brain edema, and angioedema, which limit the efficacy of this therapeutic strategy. Preclinical and clinical evidence suggests that intravenous thrombolysis generates large amounts of bradykinin, a peptide with potent pro-inflammatory, and pro-edematous effects. This tPA-triggered generation of bradykinin could participate in the deleterious effects of thrombolysis and is a potential target to improve neurological outcome in tPA-treated patients. The present review aims at summarizing current evidence linking thrombolysis, bradykinin generation, and neurovascular damage.
Collapse
Affiliation(s)
- Maxime Gauberti
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France.,Department of Diagnostic Imaging and Interventional Radiology, Centre Hospitalier Universitaire Caen Côte de Nacre, Caen, France
| | - Fanny Potzeha
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France.,Department of Clinical Research, Centre Hospitalier Universitaire Caen, Caen, France
| | - Sara Martinez de Lizarrondo
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France
| |
Collapse
|
27
|
|
28
|
Gusev EI, Martynov MY, Yasamanova AN, Nikonov AA, Markin SS, Semenov AM. Thrombolytic therapy of ischemic stroke. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:4-14. [DOI: 10.17116/jnevro20181181224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Lekoubou A, Awoumou JJ, Kengne AP. Incidence of seizure in stroke patients treated with recombinant tissue plasminogen activator: A systematic review and meta-analysis. Int J Stroke 2017; 12:923-931. [DOI: 10.1177/1747493017729239] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background Recombinant tissue plasminogen activator is the only FDA-approved thrombolytic agent for acute stroke treatment. However, there are concerns that recombinant tissue plasminogen activator may increase the risk of seizures (including early and late seizures). Aims We performed a systematic review to assess the incidence of seizures and the association of recombinant tissue plasminogen activator with seizure occurrence. Summary of review We searched major databases for articles published between 1995 and February 2016. The pooled incidence of post-stroke seizure, early seizure, late seizure, and seizures sub-types was estimated overall and by status for recombinant tissue plasminogen activator treatment, and unadjusted odds ratio used to quantify the effects of recombinant tissue plasminogen activator on post-stroke seizure occurrence. In all, 4362 stroke participants were included with 49–63% being male and median age ranging from 68 to 71 years. A total of 792 received recombinant tissue plasminogen activator. The incidence of post-stroke seizure per 1000 participants (95% CI) was 95 (31–196) overall, 113 (49–202) in recombinant tissue plasminogen activator and 169 (6–326) in non-recombinant tissue plasminogen activator-treated (all heterogeneity- p<0.0001). Incidence of early seizure per 1000 (95% CI) was 35 (27–45) overall; 34 (22–50) among recombinant tissue plasminogen activator-treated patients, and 36 (25–48) among recombinant tissue plasminogen activator naïve participants (all heterogeneity- p > 0.826). The pool incidence rate per 1000 (95% CI) of late seizure was 84 (4–263), 46 (2–145), and 212 (184–241), respectively, in the overall, the recombinant tissue plasminogen activator-treated group and non-recombinant tissue plasminogen activator-treated group (heterogeneity for overall and recombinant tissue plasminogen activator-treated group < 0.0001, non-recombinant tissue plasminogen activator naïve = 0.999). The pooled odds ratio for post-stroke seizure (recombinant tissue plasminogen activator vs. no recombinant tissue plasminogen activator) was 0.94 (95% CI: 0. 17–5.26, heterogeneity- p < 0.0001). The pooled incidence per 1000 participants (95% CI) was 30 (0–144), 17 (2–49), 16 (2–44), and 9 (0–50), respectively, for focal seizure without impairment of consciousness, focal seizure with impairment of consciousness, generalized convulsive seizure, and status epilepticus; all heterogeneity- p < 0.0003. Accompanying pooled odds ratio (recombinant tissue plasminogen activator vs. no recombinant tissue plasminogen activator) based on one study was always in favor of non-significantly lower risk in recombinant tissue plasminogen activator-treated patients (all heterogeneity- p = 1). There were insufficient data to compute pooled odds ratio for early and late seizure. Conclusions Seizures affect nearly 1 out of every 10 stroke patients with inconclusive suggestion that rates are similar in recombinant tissue plasminogen activator-treated and recombinant tissue plasminogen activator naïve patients. Large prospective studies are needed to better understand the relationship between recombinant tissue plasminogen activator and post-stroke seizure occurrence.
Collapse
Affiliation(s)
- Alain Lekoubou
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA
| | | | - André Pascal Kengne
- Department of Medicine, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council of South Africa, Cape Town, South Africa
| |
Collapse
|
30
|
Can the benefits of rtPA treatment for stroke be improved? Rev Neurol (Paris) 2017; 173:566-571. [PMID: 28797689 DOI: 10.1016/j.neurol.2017.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/02/2017] [Accepted: 07/07/2017] [Indexed: 12/14/2022]
Abstract
Tissue-type plasminogen activator (tPA) is a serine protease well known to promote fibrinolysis. This is why: its recombinant form (rtPA) can be used, either alone or combined with thrombectomy, to promote recanalization/reperfusion following ischemic stroke. However, its overall benefits are counteracted by some of its side-effects, including incomplete lysis of clots, an increased risk of hemorrhagic transformation and the possibility of neurotoxicity. Nevertheless, better understanding of the mechanisms by which tPA influences brain function and promotes its alteration may help in the design of new strategies to improve stroke therapy.
Collapse
|
31
|
Louessard M, Bardou I, Lemarchand E, Thiebaut AM, Parcq J, Leprince J, Terrisse A, Carraro V, Fafournoux P, Bruhat A, Orset C, Vivien D, Ali C, Roussel BD. Activation of cell surface GRP78 decreases endoplasmic reticulum stress and neuronal death. Cell Death Differ 2017. [PMID: 28644439 DOI: 10.1038/cdd.2017.35] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The unfolded protein response (UPR) is an endoplasmic reticulum (ER) -related stress conserved pathway that aims to protect cells from being overwhelmed. However, when prolonged, UPR activation converts to a death signal, which relies on its PERK-eIF2α branch. Overactivation of the UPR has been implicated in many neurological diseases, including cerebral ischaemia. Here, by using an in vivo thromboembolic model of stroke on transgenic ER stress-reporter mice and neuronal in vitro models of ischaemia, we demonstrate that ischaemic stress leads to the deleterious activation of the PERK branch of the UPR. Moreover, we show that the serine protease tissue-type plasminogen activator (tPA) can bind to cell surface Grp78 (78 kD glucose-regulated protein), leading to a decrease of the PERK pathway activation, thus a decrease of the deleterious factor CHOP, and finally promotes neuroprotection. Altogether, this work highlights a new role and a therapeutic potential of the chaperone protein Grp78 as a membrane receptor of tPA capable to prevent from ER stress overactivation.
Collapse
Affiliation(s)
- Morgane Louessard
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Isabelle Bardou
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Eloïse Lemarchand
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Audrey M Thiebaut
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Jérôme Parcq
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Jérôme Leprince
- Normandie Univ, UNIROUEN, INSERM, Laboratoire Différenciation et Communication Neuronale et Neuroendocrine, Plate-forme de Recherche en Imagerie Cellulaire de Normandie (PRIMACEN), Rouen, France
| | - Anne Terrisse
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Valérie Carraro
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Pierre Fafournoux
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Alain Bruhat
- INRA, UMR 1019 Nutrition Humaine, Centre de Clermont-Ferrand-Theix, Saint Genès Champanelle, France
| | - Cyrille Orset
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France.,Clinical Research Department, Medical Center, University Caen Normandy, Centre Hospitalo-Universitaire Caen Côte de Nacre, Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| | - Benoit D Roussel
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders, Caen, France
| |
Collapse
|
32
|
Luo P, Liu D, Guo L. Protecting Oligodendrocytes by Targeting Non-Glutamate Receptors as a New Therapeutic Strategy for Ischemic Stroke. Pharmacology 2017. [PMID: 28637049 DOI: 10.1159/000477939] [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: 11/19/2022]
Abstract
Ischemic stroke has many devastating effects within the brain. At the cellular level, excitotoxicity has been a popular pharmacological target for therapeutics. To date, many clinical trials have been performed with drugs that target excitatory neurotransmitter receptors, such as NMDA receptor agonists. The results, however, have been lackluster. Most efforts to understand the impacts of excitotoxicity on the brain have focused primarily on neurons, and to a lesser degree, on gliocytes as cellular targets. Recent evidence suggests that oligodendrocytes (OLGs), the myelin-forming cells in the central nervous system, are damaged by ischemia in a manner completely different from that in neurons. Whereas ischemia primarily damages neurons through overactivation of ionotropic glutamate receptors, the ischemia damage in OLGs occurs through overactivation of H+-gated transient receptor potential channels. Given the differential mechanisms of ischemic injury between neurons and OLGs, strategies to target non-glutamate receptors to prevent OLG damage/demyelination deserve greater attention in drug development. Such strategies, combined with neuroprotective measures, could provide an excellent therapeutic avenue for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Pan Luo
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | |
Collapse
|
33
|
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
|
34
|
Aging of cerebral white matter. Ageing Res Rev 2017; 34:64-76. [PMID: 27865980 DOI: 10.1016/j.arr.2016.11.006] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 10/21/2016] [Accepted: 11/04/2016] [Indexed: 12/12/2022]
Abstract
White matter (WM) occupies a large volume of the human cerebrum and is mainly composed of myelinated axons and myelin-producing glial cells. The myelinated axons within WM are the structural foundation for efficient neurotransmission between cortical and subcortical areas. Similar to neuron-enriched gray matter areas, WM undergoes a series of changes during the process of aging. WM malfunction can induce serious neurobehavioral and cognitive impairments. Thus, age-related changes in WM may contribute to the functional decline observed in the elderly. In addition, aged WM becomes more susceptible to neurological disorders, such as stroke, traumatic brain injury (TBI), and neurodegeneration. In this review, we summarize the structural and functional alterations of WM in natural aging and speculate on the underlying mechanisms. We also discuss how age-related WM changes influence the progression of various brain disorders, including ischemic and hemorrhagic stroke, TBI, Alzheimer's disease, and Parkinson's disease. Although the physiology of WM is still poorly understood relative to gray matter, WM is a rational therapeutic target for a number of neurological and psychiatric conditions.
Collapse
|
35
|
Leonetti C, Macrez R, Pruvost M, Hommet Y, Bronsard J, Fournier A, Perrigault M, Machin I, Vivien D, Clemente D, De Castro F, Maubert E, Docagne F. Tissue-type plasminogen activator exerts EGF-like chemokinetic effects on oligodendrocytes in white matter (re)myelination. Mol Neurodegener 2017; 12:20. [PMID: 28231842 PMCID: PMC5322587 DOI: 10.1186/s13024-017-0160-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 02/10/2017] [Indexed: 01/12/2023] Open
Abstract
Background The ability of oligodendrocyte progenitor cells (OPCs) to give raise to myelin forming cells during developmental myelination, normal adult physiology and post-lesion remyelination in white matter depends on factors which govern their proliferation, migration and differentiation. Tissue plasminogen activator (tPA) is a serine protease expressed in the central nervous system (CNS), where it regulates cell fate. In particular, tPA has been reported to protect oligodendrocytes from apoptosis and to facilitate the migration of neurons. Here, we investigated whether tPA can also participate in the migration of OPCs during CNS development and during remyelination after focal white matter lesion. Methods OPC migration was estimated by immunohistological analysis in spinal cord and corpus callosum during development in mice embryos (E13 to P0) and after white matter lesion induced by the stereotactic injection of lysolecithin in adult mice (1 to 21 days post injection). Migration was compared in these conditions between wild type and tPA knock-out animals. The action of tPA was further investigated in an in vitro chemokinesis assay. Results OPC migration along vessels is delayed in tPA knock-out mice during development and during remyelination. tPA enhances OPC migration via an effect dependent on the activation of epidermal growth factor receptor. Conclusion Endogenous tPA facilitates the migration of OPCs during development and during remyelination after white matter lesion by the virtue of its epidermal growth factor-like domain. Electronic supplementary material The online version of this article (doi:10.1186/s13024-017-0160-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camille Leonetti
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Richard Macrez
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Mathilde Pruvost
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Jérémie Bronsard
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Antoine Fournier
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Maxime Perrigault
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Isabel Machin
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain.,Grupo de Neuroinmuno-reparación, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Diego Clemente
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain.,Grupo de Neuroinmuno-reparación, Hospital Nacional de Parapléjicos, Toledo, Spain
| | - Fernando De Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos-SESCAM, Toledo, Spain.,Grupo de Neurobiología del Desarrollo (GNDe), Instituto Cajal, CSIC, Madrid, Spain
| | - Eric Maubert
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France
| | - Fabian Docagne
- Normandie Univ, UNICAEN, INSERM U1237, Physiology and imaging of neurological disorders (PhIND), Cyceron, Caen, 14000, France. .,Inserm, Centre Cyceron, Bvd Becquerel, BP5229, Caen Cedex, 14074, France.
| |
Collapse
|
36
|
Carter AR, McAvoy MP, Siegel JS, Hong X, Astafiev SV, Rengachary J, Zinn K, Metcalf NV, Shulman GL, Corbetta M. Differential white matter involvement associated with distinct visuospatial deficits after right hemisphere stroke. Cortex 2016; 88:81-97. [PMID: 28081452 DOI: 10.1016/j.cortex.2016.12.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 07/07/2016] [Accepted: 12/08/2016] [Indexed: 02/05/2023]
Abstract
Visuospatial attention depends on the integration of multiple processes, and people with right hemisphere lesions after a stroke may exhibit severe or no visuospatial deficits. The anatomy of core components of visuospatial attention is an area of intense interest. Here we examine the relationship between the disruption of core components of attention and lesion distribution in a heterogeneous group (N = 70) of patients with right hemisphere strokes regardless of the presence of clinical neglect. Deficits of lateralized spatial orienting, measured as the difference in reaction times for responding to visual targets in the contralesional or ipsilesional visual field, and deficits in re-orienting attention, as measured by the difference in reaction times for invalidly versus validly cued targets, were measured using a computerized spatial orienting task. Both measures were related through logistic regression and a novel ridge regression method to anatomical damage measured with magnetic resonance imaging. While many regions were common to both deficit maps, a deficit in lateralized spatial orienting was more associated with lesions in the white matter underlying the posterior parietal cortex, and middle and inferior frontal gyri. A deficit in re-orienting of attention toward unattended locations was associated with lesions in the white matter of the posterior parietal cortex, insular cortex and less so with white matter involvement of the anterior frontal lobe. An hodological analysis also supports this partial dissociation between the white matter tracts that are damaged in lateralized spatial biases versus impaired re-orienting. Our results underscore that the integrity of fronto-parietal white matter tracts is crucial for visuospatial attention and that different attention components are mediated by partially distinct neuronal substrates.
Collapse
Affiliation(s)
- Alex R Carter
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| | - Mark P McAvoy
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua S Siegel
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xin Hong
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Serguei V Astafiev
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jennifer Rengachary
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kristi Zinn
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas V Metcalf
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gordon L Shulman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Maurizio Corbetta
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
37
|
Bonaventura A, Montecucco F, Dallegri F. Update on the effects of treatment with recombinant tissue-type plasminogen activator (rt-PA) in acute ischemic stroke. Expert Opin Biol Ther 2016; 16:1323-1340. [PMID: 27548625 DOI: 10.1080/14712598.2016.1227779] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Acute ischemic stroke (AIS) represents a major cause of death and disability all over the world. The recommended therapy aims at dissolving the clot to re-establish quickly the blood flow to the brain and reduce neuronal injury. Intravenous administration of recombinant tissue-type plasminogen activator (rt-PA) is clinically used with this goal. AREAS COVERED A description of beneficial and detrimental effects of rt-PA treatment is addressed. An overview of new therapies against AIS, such as new thrombolytics, sonolysis and sonothrombolysis, endovascular procedures, and association therapies is provided. Updates on the pathophysiological process leading to intracranial hemorrhage (ICH) is also discussed. EXPERT OPINION rt-PA treatment in AIS patients is beneficial to recovery outcomes. To weaken risks and improve benefits, it might be relevant to consider: i) a definitive identification of risk factors for symptomatic ICH; ii). a better organization of the health care system to reduce time-to-treatment and enhance discharge management. The pharmacological improvement of new thrombolytic drugs (such as tenecteplase and desmoteplase) targeting harmful and maximally exploiting beneficial effects might further reduce mortality and disability in AIS.
Collapse
Affiliation(s)
- Aldo Bonaventura
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy
- b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
| | - Fabrizio Montecucco
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy
- b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
- c Centre of Excellence for Biomedical Research (CEBR) , University of Genoa , Genoa , Italy
| | - Franco Dallegri
- a First Clinic of Internal Medicine, Department of Internal Medicine , University of Genoa School of Medicine , Genoa , Italy
- b IRCCS AOU San Martino - IST, Genoa , Genoa , Italy
| |
Collapse
|
38
|
Wang Y, Liu G, Hong D, Chen F, Ji X, Cao G. White matter injury in ischemic stroke. Prog Neurobiol 2016; 141:45-60. [PMID: 27090751 PMCID: PMC5677601 DOI: 10.1016/j.pneurobio.2016.04.005] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/01/2016] [Accepted: 04/10/2016] [Indexed: 02/06/2023]
Abstract
Stroke is one of the major causes of disability and mortality worldwide. It is well known that ischemic stroke can cause gray matter injury. However, stroke also elicits profound white matter injury, a risk factor for higher stroke incidence and poor neurological outcomes. The majority of damage caused by stroke is located in subcortical regions and, remarkably, white matter occupies nearly half of the average infarct volume. Indeed, white matter is exquisitely vulnerable to ischemia and is often injured more severely than gray matter. Clinical symptoms related to white matter injury include cognitive dysfunction, emotional disorders, sensorimotor impairments, as well as urinary incontinence and pain, all of which are closely associated with destruction and remodeling of white matter connectivity. White matter injury can be noninvasively detected by MRI, which provides a three-dimensional assessment of its morphology, metabolism, and function. There is an urgent need for novel white matter therapies, as currently available strategies are limited to preclinical animal studies. Optimal protection against ischemic stroke will need to encompass the fortification of both gray and white matter. In this review, we discuss white matter injury after ischemic stroke, focusing on clinical features and tools, such as imaging, manifestation, and potential treatments. We also briefly discuss the pathophysiology of WMI and future research directions.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Gang Liu
- Department of Neurology, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China
| | - Dandan Hong
- Department of Bioengineering, University of Pittsburgh School of Engineering, United States
| | - Fenghua Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States
| | - Xunming Ji
- Department of Neurosurgery, Xuanwu Hospital, Capital University of Medicine, Beijing 100053, China.
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, United States; Geriatric Research Education and Clinical Centers, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, United States.
| |
Collapse
|
39
|
van Overbeek EC, Staals J, Knottnerus ILH, ten Cate H, van Oostenbrugge RJ. Plasma tPA-Activity and Progression of Cerebral White Matter Hyperintensities in Lacunar Stroke Patients. PLoS One 2016; 11:e0150740. [PMID: 26942412 PMCID: PMC4778794 DOI: 10.1371/journal.pone.0150740] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 02/18/2016] [Indexed: 11/18/2022] Open
Abstract
Introduction Tissue plasminogen activator (tPA)-activity and plasminogen activator inhibitor type 1 (PAI-1) antigen are considered to be haemostasis-related markers of endothelial activation and relate to presence of cerebral white matter hyperintensities (WMH) as was earlier shown in a cross-sectional study. We investigated whether tPA-activity and PAI-1 levels are associated with WMH progression in a longitudinal study. Methods In 127 first-ever lacunar stroke patients in whom baseline brain MRI and plasma levels of tPA-activity and PAI-1-antigen were available, we obtained a 2-year follow-up MRI. We assessed WMH progression by a visual WMH change scale. We determined the relationship between levels of tPA-activity and PAI-1 and WMH progression, by logistic regression analysis. Results Plasma tPA-activity was associated with periventricular WMH progression (OR 2.36, 95% CI 1.01–5.49, with correction for age and sex and baseline presence of WMH), but not with deep or any (periventricular and/or deep) WMH progression. PAI-1 levels were lower in patients with WMH progression, but these results were not significant. Conclusion We found a relationship between plasma tPA-activity and progression of periventricular WMH. More research is needed to determine whether there is a (direct) role of tPA in the development and progression of WMH.
Collapse
Affiliation(s)
- Ellen C. van Overbeek
- Department of Neurology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
- * E-mail:
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht, MUMC, Maastricht, the Netherlands
| | | | - Hugo ten Cate
- Cardiovascular Research Institute Maastricht, MUMC, Maastricht, the Netherlands
- Department of Internal Medicine, MUMC, Maastricht, the Netherlands
| | - Robert J. van Oostenbrugge
- Department of Neurology, Maastricht University Medical Centre (MUMC), Maastricht, the Netherlands
- Cardiovascular Research Institute Maastricht, MUMC, Maastricht, the Netherlands
| |
Collapse
|
40
|
Release of Matrix Metalloproteinases-2 and 9 by S-Nitrosylated Caveolin-1 Contributes to Degradation of Extracellular Matrix in tPA-Treated Hypoxic Endothelial Cells. PLoS One 2016; 11:e0149269. [PMID: 26881424 PMCID: PMC4755609 DOI: 10.1371/journal.pone.0149269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 01/30/2016] [Indexed: 11/19/2022] Open
Abstract
Intracranial hemorrhage remains the most feared complication in tissue plasminogen activator (tPA) thrombolysis for ischemic stroke. However, the underlying molecular mechanisms are still poorly elucidated. In this study, we reported an important role of caveolin-1 (Cav-1) s-nitrosylation in matrix metalloproteinase (MMP)-2 and 9 secretion from tPA-treated ischemic endothelial cells. Brain vascular endothelial cells (bEND3) were exposed to oxygen-glucose deprivation (OGD) for 2 h before adding recombinant human tPA for 6 h. This treatment induced a significant increase of MMP2 and 9 in the media of bEND3 cells and a simultaneous degradation of fibronectin and laminin β-1, the two main components of extracellular matrix (ECM). Inhibition of MMP2 and 9 with SB-3CT completely blocked the degradation of fibronectin and laminin β-1. ODG+tPA treatment led to Cav-1 shedding from bEND3 cells into the media. Notably, OGD triggered nitric oxide (NO) production and S-nitrosylationof Cav-1 (SNCav-1). Meanwhile tPA induced activation of ERK signal pathway and stimulates the secretion of SNCav-1. Pretreatment of bEND3 cells with C-PTIO (a NO scavenger) or U0126 (a specific ERK inhibitor) significantly reduced OGD-induced S-nitrosylation of Cav-1 in cells and blocked the secretion of Cav-1 and MMP2 and 9 into the media as well as the degradation of fibronectin and laminin β-1 in OGD and tPA-treated cells. These data indicate that OGD-triggered Cav-1 S-nitrosylation interacts with tPA-induced ERK activation to augment MMP2 and 9 secretion and subsequent ECM degradation, which may account for the exacerbation of ischemic blood brain barrier damage following tPA thrombolysis for ischemic stroke.
Collapse
|
41
|
Liu X, Liu J, Zhao S, Zhang H, Cai W, Cai M, Ji X, Leak RK, Gao Y, Chen J, Hu X. Interleukin-4 Is Essential for Microglia/Macrophage M2 Polarization and Long-Term Recovery After Cerebral Ischemia. Stroke 2016; 47:498-504. [PMID: 26732561 DOI: 10.1161/strokeaha.115.012079] [Citation(s) in RCA: 297] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/25/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND PURPOSE Interleukin-4 (IL-4) is a unique cytokine that may contribute to brain repair by regulating microglia/macrophage functions. Thus, we examined the effect of IL-4 on long-term recovery and microglia/macrophage polarization in 2 well-established stroke models. METHODS Transient middle cerebral artery occlusion or permanent distal middle cerebral artery occlusion was induced in wild-type and IL-4 knockout C57/BL6 mice. In a separate cohort of wild-type animals, IL-4 (60 ng/d for 7 days) or vehicle was infused into the cerebroventricle after transient middle cerebral artery occlusion. Behavioral outcomes were assessed by the Rotarod, corner, foot fault, and Morris water maze tests. Neuronal tissue loss was verified by 2 independent neuron markers. Markers of classically activated (M1) and alternatively activated (M2) microglia were assessed by real-time polymerase chain reaction, immunofluorescence, and flow cytometry. RESULTS Loss of IL-4 exacerbated sensorimotor deficits and impaired cognitive functions ≤21 days post injury. In contrast to the delayed deterioration of neurological functions, IL-4 deficiency increased neuronal tissue loss only in the acute phase (5 days) after stroke and had no impact on neuronal tissue loss 14 or 21 days post injury. Loss of IL-4 promoted expression of M1 microglia/macrophage markers and impaired expression of M2 markers at 5 and 14 days post injury. Administration of IL-4 into the ischemic brain also enhanced long-term functional recovery. CONCLUSIONS The cytokine IL-4 improves long-term neurological outcomes after stroke, perhaps through M2 phenotype induction in microglia/macrophages. These results are the first to suggest that immunomodulation with IL-4 is a promising approach to promote long-term functional recovery after stroke.
Collapse
Affiliation(s)
- Xiangrong Liu
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Jia Liu
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Shangfeng Zhao
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Haiyue Zhang
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Wei Cai
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Mengfei Cai
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Xunming Ji
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Rehana K Leak
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Yanqin Gao
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Jun Chen
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.)
| | - Xiaoming Hu
- From the Department of Neurology, University of Pittsburgh School of Medicine, PA (X.L., S.Z., H.Z., W.C., J.C., X.H.); Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, P.R. China (X.L., X.J.); State Key Laboratory of Medical Neurobiology, Institute of Brain Sciences, Fudan University, Shanghai, China (J.L., M.C., Y.G., J.C., X.H.); Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA (R.K.L.); and Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, PA (J.C., X.H.).
| |
Collapse
|
42
|
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
|
43
|
The plasminogen activation system in neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2015; 1862:395-402. [PMID: 26493446 DOI: 10.1016/j.bbadis.2015.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 01/30/2023]
Abstract
The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
44
|
Chevilley A, Lesept F, Lenoir S, Ali C, Parcq J, Vivien D. Impacts of tissue-type plasminogen activator (tPA) on neuronal survival. Front Cell Neurosci 2015; 9:415. [PMID: 26528141 PMCID: PMC4607783 DOI: 10.3389/fncel.2015.00415] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/01/2015] [Indexed: 11/18/2022] Open
Abstract
Tissue-type plasminogen activator (tPA) a serine protease is constituted of five functional domains through which it interacts with different substrates, binding proteins, and receptors. In the last years, great interest has been given to the clinical relevance of targeting tPA in different diseases of the central nervous system, in particular stroke. Among its reported functions in the central nervous system, tPA displays both neurotrophic and neurotoxic effects. How can the protease mediate such opposite functions remain unclear but several hypotheses have been proposed. These include an influence of the degree of maturity and/or the type of neurons, of the level of tPA, of its origin (endogenous or exogenous) or of its form (single chain tPA versus two chain tPA). In this review, we will provide a synthetic snapshot of our current knowledge regarding the natural history of tPA and discuss how it sustains its pleiotropic functions with focus on excitotoxic/ischemic neuronal death and neuronal survival.
Collapse
Affiliation(s)
- Arnaud Chevilley
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Flavie Lesept
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Sophie Lenoir
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Carine Ali
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Jérôme Parcq
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| | - Denis Vivien
- INSERM, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie Caen, France
| |
Collapse
|
45
|
Conformations of tissue plasminogen activator (tPA) orchestrate neuronal survival by a crosstalk between EGFR and NMDAR. Cell Death Dis 2015; 6:e1924. [PMID: 26469972 PMCID: PMC4632317 DOI: 10.1038/cddis.2015.296] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/27/2015] [Accepted: 08/17/2015] [Indexed: 01/04/2023]
Abstract
Tissue-type plasminogen activator (tPA) is a pleiotropic serine protease of the central nervous system (CNS) with reported neurotrophic and neurotoxic functions. Produced and released under its single chain form (sc), the sc-tPA can be cleaved by plasmin or kallikrein in a two chain form, tc-tPA. Although both sc-tPA and tc-tPA display a similar fibrinolytic activity, we postulated here that these two conformations of tPA (sc-tPA and tc-tPA) could differentially control the effects of tPA on neuronal survival. Using primary cultures of mouse cortical neurons, our present study reveals that sc-tPA is the only one capable to promote N-methyl-D-aspartate receptor (NMDAR)-induced calcium influx and subsequent excitotoxicity. In contrast, both sc-tPA and tc-tPA are capable to activate epidermal growth factor receptors (EGFRs), a mechanism mediating the antiapoptotic effects of tPA. Interestingly, we revealed a tPA dependent crosstalk between EGFR and NMDAR in which a tPA-dependent activation of EGFRs leads to downregulation of NMDAR signaling and to subsequent neurotrophic effects.
Collapse
|
46
|
Tissue Plasminogen Activator Expression Is Restricted to Subsets of Excitatory Pyramidal Glutamatergic Neurons. Mol Neurobiol 2015; 53:5000-12. [PMID: 26377106 DOI: 10.1007/s12035-015-9432-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022]
Abstract
Although the extracellular serine protease tissue plasminogen activator (tPA) is involved in pathophysiological processes such as learning and memory, anxiety, epilepsy, stroke, and Alzheimer's disease, information about its regional, cellular, and subcellular distribution in vivo is lacking. In the present study, we observed, in healthy mice and rats, the presence of tPA in endothelial cells, oligodendrocytes, mastocytes, and ependymocytes, but not in pericytes, microglial cells, and astrocytes. Moreover, blockage of the axo-dendritic transport unmasked tPA expression in neurons of cortical and hippocampal areas. Interestingly, combined electrophysiological recordings, single-cell reverse transcription polymerase chain reaction (RT-PCR), and immunohistological analyses revealed that the presence of tPA is restricted to subsets of excitatory pyramidal glutamatergic neurons. We further evidenced that tPA is stored in synaptobrevin-2-positive glutamatergic synaptic vesicles. Based on all these data, we propose the existence of tPA-ergic neurons in the mature brain.
Collapse
|
47
|
Lemarchand E, Maubert E, Haelewyn B, Ali C, Rubio M, Vivien D. Stressed neurons protect themselves by a tissue-type plasminogen activator-mediated EGFR-dependent mechanism. Cell Death Differ 2015; 23:123-31. [PMID: 26068590 DOI: 10.1038/cdd.2015.76] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 11/09/2022] Open
Abstract
In the central nervous system, tissue-type plasminogen activator (tPA) has been associated with both pro-death and prosurvival actions on neurons. In most cases, this has been related to exogenous tPA. In the present study, we addressed the influence of endogenous tPA. We first observed an increased transcription of tPA following either in vivo global brain ischemia in rats or in vitro oxygen glucose deprivation (OGD) on mice and rats hippocampal slices. Hippocampal slices from tPA-deficient mice were more sensitive to OGD than wild-type slices. Pharmacological approaches targeting the known receptors of tPA revealed that only the inhibition of phosphorylation of epidermal growth factor receptors (EGFRs) prevented the neuroprotective effect of endogenous tPA. This study shows that ischemic hippocampal neurons overproduce endogenous tPA as an intend to protect themselves from ischemic death, by a mechanism involving an activation of EGFRs. Thus, strategies contributing to promote either endogenous production of tPA or its associated EGFR-linked signaling pathway may have beneficial effects following brain injuries such as stroke.
Collapse
Affiliation(s)
- E Lemarchand
- INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, Bd Henri Becquerel, GIP Cyceron, Caen, France
| | - E Maubert
- INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, Bd Henri Becquerel, GIP Cyceron, Caen, France
| | - B Haelewyn
- ESRP (European Stroke Research Platform), Centre Universitaire de Ressources Biologiques (CURB), Université Caen Basse Normandie, Caen, France
| | - C Ali
- INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, Bd Henri Becquerel, GIP Cyceron, Caen, France
| | - M Rubio
- INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, Bd Henri Becquerel, GIP Cyceron, Caen, France
| | - D Vivien
- INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, Bd Henri Becquerel, GIP Cyceron, Caen, France
| |
Collapse
|
48
|
Gulati P, Muthuraman A, Kaur P. Investigation of the role of non-selective calcium channel blocker (flunarizine) on cerebral ischemic-reperfusion associated cognitive dysfunction in aged mice. Pharmacol Biochem Behav 2015; 131:26-32. [PMID: 25636603 DOI: 10.1016/j.pbb.2015.01.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 12/24/2022]
Abstract
The present study was designed to investigate the role of flunarizine (a non-selective calcium channel blocker) on cerebral ischemic-reperfusion associated cognitive dysfunction in aged mice. Bilateral carotid artery occlusion of 12min followed by reperfusion for 24h was given to induce cerebral injury in male Swiss mice. The assessment of learning & memory was performed by Morris water maze test; motor in-coordination was evaluated by rota rod, lateral push and inclined beam walking tests; cerebral infarct size was quantified by triphenyltetrazolium chloride staining. In addition, reduced glutathione (GSH), total calcium and acetylcholinesterase (AChE) activity were also estimated in aged brain tissue. Donepezil treated group served as a positive control in this study. Ischemia reperfusion (I/R) injury produced significant increase in cerebral infarct size. A significant loss of memory along with impairment of motor performance was also noted. Further, I/R injury also produced significant increase in levels of total calcium, AChE activity and decrease in GSH levels. Pretreatment of flunarizine significantly attenuated I/R induced infarct size, behavioral and biochemical changes. Hence, it may be concluded that, a non-selective calcium channel blocker can be useful in I/R associated cognitive dysfunction due to its anti-oxidant, anti-infarct and modulatory actions of neurotransmitters & calcium channels.
Collapse
Affiliation(s)
- Puja Gulati
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala 147002, Punjab, India
| | - Arunachalam Muthuraman
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala 147002, Punjab, India; Department of Pharmacology, Neuropharmacology Division, Akal Pharmacology & Toxicology Research Centre, Akal College of Pharmacy & Technical Education (ACPTE), Masuana Sahib, Sangrur 148001, Punjab, India.
| | - Parneet Kaur
- Department of Pharmacology, Neuropharmacology Division, Akal Pharmacology & Toxicology Research Centre, Akal College of Pharmacy & Technical Education (ACPTE), Masuana Sahib, Sangrur 148001, Punjab, India
| |
Collapse
|
49
|
Docagne F, Parcq J, Lijnen R, Ali C, Vivien D. Understanding the Functions of Endogenous and Exogenous Tissue-Type Plasminogen Activator During Stroke. Stroke 2015; 46:314-20. [DOI: 10.1161/strokeaha.114.006698] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Fabian Docagne
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Jérôme Parcq
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Roger Lijnen
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Carine Ali
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Denis Vivien
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| |
Collapse
|
50
|
A Smoothened receptor agonist is neuroprotective and promotes regeneration after ischemic brain injury. Cell Death Dis 2014; 5:e1481. [PMID: 25341035 PMCID: PMC4649529 DOI: 10.1038/cddis.2014.446] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 09/08/2014] [Accepted: 09/09/2014] [Indexed: 12/26/2022]
Abstract
Ischemic stroke occurs as a result of blood supply interruption to the brain causing tissue degeneration, patient disabilities or death. Currently, treatment of ischemic stroke is limited to thrombolytic therapy with a narrow time window of administration. The sonic hedgehog (Shh) signaling pathway has a fundamental role in the central nervous system development, but its impact on neural cell survival and tissue regeneration/repair after ischemic stroke has not been well investigated. Here we report the neuroprotective properties of a small-molecule agonist of the Shh co-receptor Smoothened, purmorphamine (PUR), in the middle cerebral artery occlusion model of ischemic stroke. We found that intravenous administration of PUR at 6 h after injury was neuroprotective and restored neurological deficit after stroke. PUR promoted a transient upregulation of tissue-type plasminogen activator in injured neurons, which was associated with a reduction of apoptotic cell death in the ischemic cortex. We also observed a decrease in blood–brain barrier permeability after PUR treatment. At 14 d postinjury, attenuation of inflammation and reactive astrogliosis was found in PUR-treated animals. PUR increased the number of newly generated neurons in the peri-infarct and infarct area and promoted neovascularization in the ischemic zone. Notably, PUR treatment did not significantly alter the ischemia-induced level of Gli1, a Shh target gene of tumorigenic potential. Thus our study reports a novel pharmacological approach for postischemic treatment using a small-molecule Shh agonist, providing new insights into hedgehog signaling-mediated mechanisms of neuroprotection and regeneration after stroke.
Collapse
|