1
|
Yepes M. Reprint of: Fibrinolytic and Non-fibrinolytic Roles of Tissue-type Plasminogen Activator in the Ischemic Brain. Neuroscience 2024; 550:21-29. [PMID: 38964373 DOI: 10.1016/j.neuroscience.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/06/2023] [Indexed: 07/06/2024]
Abstract
The neurovascular unit (NVU) is assembled by endothelial cells (ECs) and pericytes, and encased by a basement membrane (BM) surveilled by microglia and surrounded by perivascular astrocytes (PVA), which in turn are in contact with synapses. Cerebral ischemia induces the rapid release of the serine proteinase tissue-type plasminogen activator (tPA) from endothelial cells, perivascular astrocytes, microglia and neurons. Owning to its ability to catalyze the conversion of plasminogen into plasmin, in the intravascular space tPA functions as a fibrinolytic enzyme. In contrast, the release of astrocytic, microglial and neuronal tPA have a plethora of effects that not always require the generation of plasmin. In the ischemic brain tPA increases the permeability of the NVU, induces microglial activation, participates in the recycling of glutamate, and has various effects on neuronal survival. These effects are mediated by different receptors, notably subunits of the N-methyl-D-aspartate receptor (NMDAR) and the low-density lipoprotein receptor-related protein-1 (LRP-1). Here we review data on the role of tPA in the NVU under non-ischemic and ischemic conditions, and analyze how this knowledge may lead to the development of potential strategies for the treatment of acute ischemic stroke patients.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, USA; Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
2
|
Yepes M. Fibrinolytic and Non-fibrinolytic Roles of Tissue-type Plasminogen Activator in the Ischemic Brain. Neuroscience 2024; 542:69-80. [PMID: 37574107 DOI: 10.1016/j.neuroscience.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/03/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
The neurovascular unit (NVU) is assembled by endothelial cells (ECs) and pericytes, and encased by a basement membrane (BM) surveilled by microglia and surrounded by perivascular astrocytes (PVA), which in turn are in contact with synapses. Cerebral ischemia induces the rapid release of the serine proteinase tissue-type plasminogen activator (tPA) from endothelial cells, perivascular astrocytes, microglia and neurons. Owning to its ability to catalyze the conversion of plasminogen into plasmin, in the intravascular space tPA functions as a fibrinolytic enzyme. In contrast, the release of astrocytic, microglial and neuronal tPA have a plethora of effects that not always require the generation of plasmin. In the ischemic brain tPA increases the permeability of the NVU, induces microglial activation, participates in the recycling of glutamate, and has various effects on neuronal survival. These effects are mediated by different receptors, notably subunits of the N-methyl-D-aspartate receptor (NMDAR) and the low-density lipoprotein receptor-related protein-1 (LRP-1). Here we review data on the role of tPA in the NVU under non-ischemic and ischemic conditions, and analyze how this knowledge may lead to the development of potential strategies for the treatment of acute ischemic stroke patients.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University, Atlanta, GA, USA; Division of Neuropharmacology and Neurologic Diseases, Emory Primate Research Center, Atlanta, GA, USA; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
3
|
Smart C, Mitchell A, McCutcheon F, Medcalf RL, Thiele A. Tissue-type plasminogen activator induces conditioned receptive field plasticity in the mouse auditory cortex. iScience 2023; 26:105947. [PMID: 36711245 PMCID: PMC9874071 DOI: 10.1016/j.isci.2023.105947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/13/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
Tissue-type plasminogen activator (tPA) is a serine protease that is expressed in various compartments in the brain. It is involved in neuronal plasticity, learning and memory, and addiction. We evaluated whether tPA, exogenously applied, could influence neuroplasticity within the mouse auditory cortex. We used a frequency-pairing paradigm to determine whether neuronal best frequencies shift following the pairing protocol. tPA administration significantly affected the best frequency after pairing, whereby this depended on the pairing frequency relative to the best frequency. When the pairing frequency was above the best frequency, tPA caused a best frequency shift away from the conditioned frequency. tPA significantly widened auditory tuning curves. Our data indicate that regional changes in proteolytic activity within the auditory cortex modulate the fine-tuning of auditory neurons, supporting the function of tPA as a modulator of neuronal plasticity.
Collapse
Affiliation(s)
- Caitlin Smart
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Anna Mitchell
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Fiona McCutcheon
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Alexander Thiele
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
4
|
Plasminogen Activators in Neurovascular and Neurodegenerative Disorders. Int J Mol Sci 2021; 22:ijms22094380. [PMID: 33922229 PMCID: PMC8122722 DOI: 10.3390/ijms22094380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
The neurovascular unit (NVU) is a dynamic structure assembled by endothelial cells surrounded by a basement membrane, pericytes, astrocytes, microglia and neurons. A carefully coordinated interplay between these cellular and non-cellular components is required to maintain normal neuronal function, and in line with these observations, a growing body of evidence has linked NVU dysfunction to neurodegeneration. Plasminogen activators catalyze the conversion of the zymogen plasminogen into the two-chain protease plasmin, which in turn triggers a plethora of physiological events including wound healing, angiogenesis, cell migration and inflammation. The last four decades of research have revealed that the two mammalian plasminogen activators, tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), are pivotal regulators of NVU function during physiological and pathological conditions. Here, we will review the most relevant data on their expression and function in the NVU and their role in neurovascular and neurodegenerative disorders.
Collapse
|
5
|
Abstract
Dementia is a clinical syndrome that affects approximately 47 million people worldwide and is characterized by progressive and irreversible decline of cognitive, behavioral and sesorimotor functions. Alzheimer's disease (AD) accounts for approximately 60-80% of all cases of dementia, and neuropathologically is characterized by extracellular deposits of insoluble amyloid-β (Aβ) and intracellular aggregates of hyperphosphorylated tau. Significantly, although for a long time it was believed that the extracellular accumulation of Aβ was the culprit of the symptoms observed in these patients, more recent studies have shown that cognitive decline in people suffering this disease is associated with soluble Aβ-induced synaptic dysfunction instead of the formation of insoluble Aβ-containing extracellular plaques. These observations are translationally relevant because soluble Aβ-induced synaptic dysfunction is an early event in AD that precedes neuronal death, and thus is amenable to therapeutic interventions to prevent cognitive decline before the progression to irreversible brain damage. The plasminogen activating (PA) system is an enzymatic cascade that triggers the degradation of fibrin by catalyzing the conversion of plasminogen into plasmin via two serine proteinases: tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). Experimental evidence reported over the last three decades has shown that tPA and uPA play a role in the pathogenesis of AD. However, these studies have focused on the ability of these plasminogen activators to trigger plasmin-induced cleavage of insoluble Aβ-containing extracellular plaques. In contrast, recent evidence indicates that activity-dependent release of uPA from the presynaptic terminal of cerebral cortical neurons protects the synapse from the deleterious effects of soluble Aβ via a mechanism that does not require plasmin generation or the cleavage of Aβ fibrils. Below we discuss the role of the PA system in the pathogenesis of AD and the translational relevance of data published to this date.
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology, Emory University School of Medicine; Department of Neurology, Veterans Affairs Medical Center; Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| |
Collapse
|
6
|
Yepes M. The Plasminogen Activation System Promotes Neurorepair in the Ischemic Brain. Curr Drug Targets 2020; 20:953-959. [PMID: 30539695 PMCID: PMC6700753 DOI: 10.2174/1389450120666181211144550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/26/2022]
Abstract
The plasminogen activation (PA) system was originally thought to exclusively promote the degradation of fibrin by catalyzing the conversion of plasminogen into plasmin via two serine proteinases: tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). However, experimental evidence accumulated over the last 30 years indicates that tPA and uPA are also found in the central nervous system (CNS), where they have a plethora of functions that not always require plasmin generation or fibrin degradation. For example, plasminogen-dependent and - independent effects of tPA and uPA play a central role in the pathophysiological events that underlie one of the leading causes of mortality and disability in the world: cerebral ischemia. Indeed, recent work indicates that while the rapid release of tPA from the presynaptic compartment following the onset of cerebral ischemia protects the synapse from the deleterious effects of the ischemic injury, the secretion of uPA and its binding to its receptor (uPAR) during the recovery phase promotes the repair of synapses that have been lost to the acute ischemic insult. This restorative role of uPA has high translational significance because to this date there is no effective approach to induce neurorepair in the ischemic brain. Here we will discuss recent evidence that bridges the gap between basic research in the field of the PA system and the bedside of ischemic stroke patients, indicating that uPA and uPAR are potential targets for the development of therapeutic strategies to promote neurological recovery among ischemic stroke survivors.
Collapse
Affiliation(s)
- Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center; Atlanta, GA, United States.,Department of Neurology & Center for Neurodegenerative Disease, Emory University School of Medicine; Atlanta, GA, United States.,Department of Neurology, Veterans Affairs Medical Center; Atlanta, GA, United States
| |
Collapse
|
7
|
White S, Lin L, Hu K. NF-κB and tPA Signaling in Kidney and Other Diseases. Cells 2020; 9:E1348. [PMID: 32485860 PMCID: PMC7348801 DOI: 10.3390/cells9061348] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023] Open
Abstract
The activation of the nuclear factor-κB (NF-κB) pathway plays a central role in the initiation and progression of inflammation, which contributes to the pathogenesis and progression of various human diseases including kidney, brain, and other diseases. Tissue plasminogen activator (tPA), a serine protease regulating homeostasis of blood coagulation, fibrinolysis, and matrix degradation, has been shown to act as a cytokine to trigger profound receptor-mediated intracellular events, modulate the NF-κB pathway, and mediate organ dysfunction and injury. In this review, we focus on the current understanding of NF-κB and tPA signaling in the development and progression of kidney disease. Their roles in the nervous and cardiovascular system are also briefly discussed.
Collapse
Affiliation(s)
| | - Ling Lin
- Nephrology Research Program, Department of Medicine, Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| | - Kebin Hu
- Nephrology Research Program, Department of Medicine, Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA;
| |
Collapse
|
8
|
Parmer RJ, Gong Y, Yoo SH, Miles LA. Neuroendocrine Targeting of Tissue Plasminogen Activator (t-PA). JOURNAL OF NEUROLOGICAL DISORDERS & STROKE 2020; 7:1153. [PMID: 32549050 PMCID: PMC7297194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
t-PA has a widespread neuroendocrine distribution including prominent expression in chromaffin cells of the sympathoadrenal system. Chromaffin cell t-PA is sorted into catecholamine storage vesicles and co-released with catecholamines in response to sympathoadrenal activation, suggesting that catecholamine storage vesicles may serve as a reservoir for the rapid release of t-PA. Chromogranin A (CgA), a major core protein in secretory vesicles throughout the neuroendocrine system, may play a crucial role in targeting proteins into the regulated secretory pathway, by forming aggregated "granin" complexes to which other proteins destined for the regulated secretory vesicle bind and become separated from constitutively secreted proteins in the trans-Golgi network (TGN). Formation of such complexes is facilitated by conditions of the TGN (low pH, high Ca+2). We tested the hypothesis that t-PA interacts specifically with CgA and that this interaction is enhanced under conditions of the TGN. Immobilized t-PA was incubated with 125I-CgA. t-PA interacted specifically and saturably with CgA and the interaction was domain-specific, mediated by the EGF/finger and kringle 1 domains of t-PA and by a specific internal hydrophilic domain within CgA (KERTHQQKKHSSYEDELSEVL) as assessed by antibody and peptide competition studies. The interaction of t-PA with aggregated CgA complexes may play a role in the targeting of t-PA and its release from neurosecretory cells. These results may have broad implications for the regulation of local neurosecretory cell plasminogen activation under both normal physiological conditions and pathological conditions including cerebral ischemia.
Collapse
Affiliation(s)
- Robert J. Parmer
- Department of Medicine, University of California, USA
- Veterans Administration San Diego Healthcare System, San Diego, CA, USA
| | - Yun Gong
- Department of Pathology, M.D. Anderson Cancer Center, USA
| | | | - Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, USA
| |
Collapse
|
9
|
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
|
10
|
Zhang F, Luo J, Zhu X. Ketamine ameliorates depressive-like behaviors by tPA-mediated conversion of proBDNF to mBDNF in the hippocampus of stressed rats. Psychiatry Res 2018; 269:646-651. [PMID: 30216916 DOI: 10.1016/j.psychres.2018.08.075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 07/26/2018] [Accepted: 08/17/2018] [Indexed: 12/29/2022]
Abstract
Some studies have indicated that ketamine has a rapid antidepressant effects, but the underlying molecular mechanism is still unclear. Researchers have found that mature brain-derived neurotrophic factor (mBDNF) and its precursor proBDNF are related to depression; they elicit opposite effects on cellular functions. It is clear that tissue plasminogen activator (tPA) is a key regulatory element in the conversion of proBDNF to mBDNF. The chronic unpredicted mild stress (CUMS) procedure is a classical and reliable method to establish the model of depression. This study found that sucrose preference and locomotor activity were both reduced in CUMS-treated rats while were increased in those who were injected with ketamine. The hippocampal proBDNF/mBDNF ratio was downregulated after ketamine treatment in those rats, together with an increased level of tPA in the hippocampus. However, tPA activity was unaltered after ketamine intraperitoneal injection. Intrahippocampal injection of active plasminogen activator inhibitor-1 (inhibitor of tPA) before ketamine treatment reversed the antidepressant effects and upregulated the proBDNF/mBDNF ratio. The results of this study suggest that the antidepressant action induced by ketamine may be related to tPA-mediated conversion of proBDNF to mBDNF in the hippocampus.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Anesthesiology, The People's Hospital of Jianyang City, Chengdu 610000, China
| | - Jie Luo
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xianlin Zhu
- Department of Anesthesiology, The Central Hospital of Enshi Autonomous Prefecture, Enshi 445000, China.
| |
Collapse
|
11
|
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
|
12
|
Wilhelm CJ, Hashimoto JG, Roberts ML, Zhang X, Goeke CM, Bloom SH, Guizzetti M. Plasminogen activator system homeostasis and its dysregulation by ethanol in astrocyte cultures and the developing brain. Neuropharmacology 2018; 138:193-209. [PMID: 29885422 PMCID: PMC6310223 DOI: 10.1016/j.neuropharm.2018.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 05/29/2018] [Accepted: 06/04/2018] [Indexed: 10/30/2022]
Abstract
In utero alcohol exposure can cause fetal alcohol spectrum disorders (FASD), characterized by structural brain abnormalities and long-lasting behavioral and cognitive dysfunction. Neuronal plasticity is affected by in utero alcohol exposure and can be modulated by extracellular proteolysis. Plasmin is a major extracellular serine-protease whose activation is tightly regulated by the plasminogen activator (PA) system. In the present study we explored the effect of ethanol on the expression of the main components of the brain PA system in sex-specific cortical astrocyte primary cultures in vitro and in the cortex and hippocampus of post-natal day (PD) 9 male and female rats. We find that ethanol alters the PA system in astrocytes and in the developing brain. In particular, the expression of tissue-type PA (tPA), encoded by the gene Plat, is consistently upregulated by ethanol in astrocytes in vitro and in the cortex and hippocampus in vivo. Astrocytes exhibit endogenous plasmin activity that is increased by ethanol and recombinant tPA and inhibited by tPA silencing. We also find that tPA is expressed by astrocytes of the developing cortex and hippocampus in vivo. All components of the PA system investigated, with the exception of Neuroserpin/Serpini1, are expressed at higher levels in astrocyte cultures than in the developing brain, suggesting that astrocytes are major producers of these proteins in the brain. In conclusion, astrocyte PA system may play a major role in the modulation of neuronal plasticity; ethanol-induced upregulation of tPA levels and plasmin activity may be responsible for altered neuronal plasticity in FASD.
Collapse
Affiliation(s)
- Clare J Wilhelm
- VA Portland Health Care System, Portland, OR, 97239, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Joel G Hashimoto
- VA Portland Health Care System, Portland, OR, 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | | | | | - Calla M Goeke
- VA Portland Health Care System, Portland, OR, 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | | | - Marina Guizzetti
- VA Portland Health Care System, Portland, OR, 97239, USA; Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
13
|
Yepes M. Urokinase-type Plasminogen Activator Promotes Synaptic Recovery in the Ischemic Brain. JOURNAL OF TRANSLATIONAL NEUROSCIENCES 2018; 3:3. [PMID: 29938252 PMCID: PMC6013270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Synaptic dysfunction underlies the development of neurological impairment following an acute ischemic stroke. Unfortunately, to this date there is no therapeutic approach to protect and repair the synapse that has suffered an ischemic injury. However, recent research with in vitro models of hypoxia, in vivo models of cerebral ischemia and different neuroradiological techniques has revealed that during the recovery phase from a hypoxic injury neurons release the serine proteinase urokinase-type plasminogen activator (uPA) and astrocytes recruit its receptor (uPAR) to their plasma membrane; and that binding of neuronal uPA to astrocytic uPAR promotes the recovery of the "tripartite synapse" that has suffered an acute ischemic injury. The translational relevance of these findings is underscored by the fact that intravenous treatment with recombinant uPA promotes synaptic recovery and functional improvement following an acute ischemic stroke.
Collapse
Affiliation(s)
- Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA,Department of Neurology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA,Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA,Corresponding author: Manuel Yepes, , MD, Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, 954 Gatewood Road-NE, Atlanta, GA 30329-4208, USA. Tel: (404) 712 8358, Fax: (404) 727 3728
| |
Collapse
|
14
|
Gerashchenko D, Pasumarthi RK, Kilduff TS. Plasticity-Related Gene Expression During Eszopiclone-Induced Sleep. Sleep 2017; 40:3866746. [PMID: 28605546 DOI: 10.1093/sleep/zsx098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Study Objectives Experimental evidence suggests that restorative processes depend on synaptic plasticity changes in the brain during sleep. We used the expression of plasticity-related genes to assess synaptic plasticity changes during drug-induced sleep. Methods We first characterized sleep induced by eszopiclone in mice during baseline conditions and during the recovery from sleep deprivation. We then compared the expression of 18 genes and two miRNAs critically involved in synaptic plasticity in these mice. Gene expression was assessed in the cerebral cortex and hippocampus by the TaqMan reverse transcription polymerase chain reaction and correlated with sleep parameters. Results Eszopiclone reduced the latency to nonrapid eye movement (NREM) sleep and increased NREM sleep amounts. Eszopiclone had no effect on slow wave activity (SWA) during baseline conditions but reduced the SWA increase during recovery sleep (RS) after sleep deprivation. Gene expression analyses revealed three distinct patterns: (1) four genes had higher expression either in the cortex or hippocampus in the group of mice with increased amounts of wakefulness; (2) a large proportion of plasticity-related genes (7 out of 18 genes) had higher expression during RS in the cortex but not in the hippocampus; and (3) six genes and the two miRNAs showed no significant changes across conditions. Even at a relatively high dose (20 mg/kg), eszopiclone did not reduce the expression of plasticity-related genes during RS period in the cortex. Conclusions These results indicate that gene expression associated with synaptic plasticity occurs in the cortex in the presence of a hypnotic medication.
Collapse
Affiliation(s)
| | - Ravi K Pasumarthi
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| |
Collapse
|
15
|
Jeanneret V, Wu F, Merino P, Torre E, Diaz A, Cheng L, Yepes M. Tissue-type Plasminogen Activator (tPA) Modulates the Postsynaptic Response of Cerebral Cortical Neurons to the Presynaptic Release of Glutamate. Front Mol Neurosci 2016; 9:121. [PMID: 27881952 PMCID: PMC5101231 DOI: 10.3389/fnmol.2016.00121] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/31/2016] [Indexed: 01/31/2023] Open
Abstract
Tissue-type plasminogen activator (tPA) is a serine proteinase released by the presynaptic terminal of cerebral cortical neurons following membrane depolarization (Echeverry et al., 2010). Recent studies indicate that the release of tPA triggers the synaptic vesicle cycle and promotes the exocytosis (Wu et al., 2015) and endocytic retrieval (Yepes et al., 2016) of glutamate-containing synaptic vesicles. Here we used electron microscopy, proteomics, quantitative phosphoproteomics, biochemical analyses with extracts of the postsynaptic density (PSD), and an animal model of cerebral ischemia with mice overexpressing neuronal tPA to study whether the presynaptic release of tPA also has an effect on the postsynaptic terminal. We found that tPA has a bidirectional effect on the composition of the PSD of cerebral cortical neurons that is independent of the generation of plasmin and the presynaptic release of glutamate, but depends on the baseline level of neuronal activity and the extracellular concentrations of calcium (Ca2+). Accordingly, in neurons that are either inactive or incubated with low Ca2+ concentrations tPA induces phosphorylation and accumulation in the PSD of the Ca2+/calmodulin-dependent protein kinase IIα (pCaMKIIα), followed by pCaMKIIα-mediated phosphorylation and synaptic recruitment of GluR1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. In contrast, in neurons with previously increased baseline levels of pCaMKIIα in the PSD due to neuronal depolarization in vivo or incubation with high concentrations of either Ca2+ or glutamate in vitro, tPA induces pCaMKIIα and pGluR1 dephosphorylation and their subsequent removal from the PSD. We found that these effects of tPA are mediated by synaptic N-methyl-D-aspartate (NMDA) receptors and cyclin-dependent kinase 5 (Cdk5)-induced phosphorylation of the protein phosphatase 1 (PP1) at T320. Our data indicate that by regulating the pCaMKIIα/PP1 balance in the PSD tPA acts as a homeostatic regulator of the postsynaptic response of cerebral cortical neurons to the presynaptic release of glutamate.
Collapse
Affiliation(s)
- Valerie Jeanneret
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine Atlanta, GA, USA
| | - Fang Wu
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine Atlanta, GA, USA
| | - Paola Merino
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine Atlanta, GA, USA
| | - Enrique Torre
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine Atlanta, GA, USA
| | - Ariel Diaz
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine Atlanta, GA, USA
| | - Lihong Cheng
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine Atlanta, GA, USA
| | - Manuel Yepes
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of MedicineAtlanta, GA, USA; Department of Neurology, Veterans Affairs Medical CenterAtlanta, GA, USA
| |
Collapse
|
16
|
Medcalf RL, Lawrence DA. Editorial: The Role of the Plasminogen Activating System in Neurobiology. Front Cell Neurosci 2016; 10:222. [PMID: 27757075 PMCID: PMC5048060 DOI: 10.3389/fncel.2016.00222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/09/2016] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robert L Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University Melbourne, VIC, Australia
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
17
|
Dahl LCM, Nasa Z, Chung J, Niego B, Tarlac V, Ho H, Galle A, Petratos S, Lee JY, Alderuccio F, Medcalf RL. The Influence of Differentially Expressed Tissue-Type Plasminogen Activator in Experimental Autoimmune Encephalomyelitis: Implications for Multiple Sclerosis. PLoS One 2016; 11:e0158653. [PMID: 27427941 PMCID: PMC4948890 DOI: 10.1371/journal.pone.0158653] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/17/2016] [Indexed: 12/21/2022] Open
Abstract
Tissue type plasminogen activator (t-PA) has been implicated in the development of multiple sclerosis (MS) and in rodent models of experimental autoimmune encephalomyelitis (EAE). We show that levels of t-PA mRNA and activity are increased ~4 fold in the spinal cords of wild-type mice that are mice subjected to EAE. This was also accompanied with a significant increase in the levels of pro-matrix metalloproteinase 9 (pro-MMP-9) and an influx of fibrinogen. We next compared EAE severity in wild-type mice, t-PA-/- mice and T4+ transgenic mice that selectively over-express (~14-fold) mouse t-PA in neurons of the central nervous system. Our results confirm that t-PA deficient mice have an earlier onset and more severe form of EAE. T4+ mice, despite expressing higher levels of endogenous t-PA, manifested a similar rate of onset and neurological severity of EAE. Levels of proMMP-9, and extravasated fibrinogen in spinal cord extracts were increased in mice following EAE onset regardless of the absence or over-expression of t-PA wild-type. Interestingly, MMP-2 levels also increased in spinal cord extracts of T4+ mice following EAE, but not in the other genotypes. Hence, while the absence of t-PA confers a more deleterious form of EAE, neuronal over-expression of t-PA does not overtly protect against this condition with regards to symptom onset or severity of EAE.
Collapse
Affiliation(s)
- Lisa CM Dahl
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Zeyad Nasa
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - JieYu Chung
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Be’eri Niego
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Volga Tarlac
- Van Cleef Roet Centre for Nervous Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Heidi Ho
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Adam Galle
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Melbourne, 3004, Victoria, Australia
| | - Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Melbourne, 3004, Victoria, Australia
| | - Frank Alderuccio
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
- * E-mail:
| |
Collapse
|
18
|
Wu F, Torre E, Cuellar-Giraldo D, Cheng L, Yi H, Bichler EK, García PS, Yepes M. Tissue-type plasminogen activator triggers the synaptic vesicle cycle in cerebral cortical neurons. J Cereb Blood Flow Metab 2015; 35:1966-76. [PMID: 26126868 PMCID: PMC4671117 DOI: 10.1038/jcbfm.2015.155] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/28/2015] [Accepted: 06/02/2015] [Indexed: 12/14/2022]
Abstract
The active zone (AZ) is a thickening of the presynaptic membrane where exocytosis takes place. Chemical synapses contain neurotransmitter-loaded synaptic vesicles (SVs) that at rest are tethered away from the synaptic release site, but after the presynaptic inflow of Ca(+2) elicited by an action potential translocate to the AZ to release their neurotransmitter load. We report that tissue-type plasminogen activator (tPA) is stored outside the AZ of cerebral cortical neurons, either intermixed with small clear-core vesicles or in direct contact with the presynaptic membrane. We found that cerebral ischemia-induced release of neuronal tPA, or treatment with recombinant tPA, recruits the cytoskeletal protein βII-spectrin to the AZ and promotes the binding of SVs to βII-spectrin, enlarging the population of SVs in proximity to the synaptic release site. This effect does not require the generation of plasmin and is followed by the recruitment of voltage gated calcium channels (VGCC) to the presynaptic terminal that leads to Ca(+2)-dependent synapsin I phosphorylation, freeing SVs to translocate to the AZ to deliver their neurotransmitter load. Our studies indicate that tPA activates the SV cycle and induces the structural and functional changes in the synapse that are required for successful neurotransmission.
Collapse
Affiliation(s)
- Fang Wu
- Department of Neurology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Enrique Torre
- Department of Neurology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David Cuellar-Giraldo
- Department of Neurology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lihong Cheng
- Department of Neurology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hong Yi
- Apkarian Integrated Electron Microscopy Core, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Edyta K Bichler
- Department of Anesthesiology, Veterans Affairs Medical Center, Atlanta, Georgia, USA.,Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Paul S García
- Department of Anesthesiology, Veterans Affairs Medical Center, Atlanta, Georgia, USA.,Department of Anesthesiology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Manuel Yepes
- Department of Neurology, Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia, USA.,Department of Neurology, Veterans Affairs Medical Center, Atlanta, Georgia, USA
| |
Collapse
|
19
|
Stefanitsch C, Lawrence ALE, Olverling A, Nilsson I, Fredriksson L. tPA Deficiency in Mice Leads to Rearrangement in the Cerebrovascular Tree and Cerebroventricular Malformations. Front Cell Neurosci 2015; 9:456. [PMID: 26648843 PMCID: PMC4663266 DOI: 10.3389/fncel.2015.00456] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/09/2015] [Indexed: 12/02/2022] Open
Abstract
The serine protease tissue-type plasminogen activator (tPA) is used as a thrombolytic agent in the management of ischemic stroke, but concerns for hemorrhagic conversion greatly limits the number of patients that receive this treatment. It has been suggested that the bleeding complications associated with thrombolytic tPA may be due to unanticipated roles of tPA in the brain. Recent work has suggested tPA regulation of neurovascular barrier integrity, mediated via platelet derived growth factor (PDGF)-C/PDGF receptor-α (PDGFRα) signaling, as a possible molecular mechanism affecting the outcome of stroke. To better understand the role of tPA in neurovascular regulation we conducted a detailed analysis of the cerebrovasculature in brains from adult tPA deficient (tPA−/−) mice. Our analysis demonstrates that life-long deficiency of tPA is associated with rearrangements in the cerebrovascular tree, including a reduction in the number of vascular smooth-muscle cell covered, large diameter, vessels and a decrease in vessel-associated PDGFRα expression as compared to wild-type (WT) littermate controls. In addition, we found that ablation of tPA results in an increased number of ERG-positive endothelial cells and increased junctional localization of the tight junction protein ZO1. This is intriguing since ERG is an endothelial transcription factor implicated in regulation of vascular integrity. Based on these results, we propose that the protection of barrier properties seen utilizing these tPA−/− mice might be due, at least in part, to these cerebrovascular rearrangements. In addition, we found that tPA−/− mice displayed mild cerebral ventricular malformations, a feature previously associated with ablation of PDGF-C, thereby providing an in vivo link between tPA and PDGF signaling in central nervous system (CNS) development. Taken together, the data presented here will advance our understanding of the role of tPA within the CNS and in regulation of cerebrovascular permeability.
Collapse
Affiliation(s)
- Christina Stefanitsch
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden
| | - Anna-Lisa E Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School Ann Arbor, MI, USA
| | - Anna Olverling
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden
| | - Ingrid Nilsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden
| | - Linda Fredriksson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet Stockholm, Sweden ; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School Ann Arbor, MI, USA
| |
Collapse
|
20
|
The Role of Proteases in Hippocampal Synaptic Plasticity: Putting Together Small Pieces of a Complex Puzzle. Neurochem Res 2015; 41:156-82. [DOI: 10.1007/s11064-015-1752-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 12/17/2022]
|
21
|
Lee TW, Tsang VWK, Birch NP. Physiological and pathological roles of tissue plasminogen activator and its inhibitor neuroserpin in the nervous system. Front Cell Neurosci 2015; 9:396. [PMID: 26528129 PMCID: PMC4602146 DOI: 10.3389/fncel.2015.00396] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/22/2015] [Indexed: 12/03/2022] Open
Abstract
Although its roles in the vascular space are most well-known, tissue plasminogen activator (tPA) is widely expressed in the developing and adult nervous system, where its activity is believed to be regulated by neuroserpin, a predominantly brain-specific member of the serpin family of protease inhibitors. In the normal physiological state, tPA has been shown to play roles in the development and plasticity of the nervous system. Ischemic damage, however, may lead to excess tPA activity in the brain and this is believed to contribute to neurodegeneration. In this article, we briefly review the physiological and pathological roles of tPA in the nervous system, which includes neuronal migration, axonal growth, synaptic plasticity, neuroprotection and neurodegeneration, as well as a contribution to neurological disease. We summarize tPA's multiple mechanisms of action and also highlight the contributions of the inhibitor neuroserpin to these processes.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand
| | - Vicky W K Tsang
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand
| | - Nigel P Birch
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand ; Brain Research New Zealand, Rangahau Roro Aotearoa Auckland, New Zealand
| |
Collapse
|
22
|
Yepes M. Tissue-type plasminogen activator is a neuroprotectant in the central nervous system. Front Cell Neurosci 2015; 9:304. [PMID: 26347605 PMCID: PMC4538299 DOI: 10.3389/fncel.2015.00304] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/27/2015] [Indexed: 01/04/2023] Open
Abstract
Tissue-type plasminogen activator (tPA) is a serine proteinase found not only in the intravascular space but also in a well-defined sub-set of neurons in the brain. tPA is rapidly released from neurons after either exposure to hypoxia or hypoglycemia in vitro, or the induction of cerebral ischemia in vivo. It has been proposed that tPA has a neurotoxic effect in the ischemic brain. However, recent evidence indicate that once released into the synaptic cleft tPA activates specific cell signaling pathways that promote the detection and adaptation to metabolic stress. More specifically, the non-proteolytic interaction of tPA with N-methyl-D-aspartate receptors (NMDARs) and a member of the low-density lipoprotein receptor (LDLR) family in dendritic spines activates the mammalian target of rapamycin (mTOR) pathway that adapts cellular processes to the availability of energy and metabolic resources. TPA-induced mTOR activation in neurons leads to hypoxia-inducible factor 1α (HIF-1α) accumulation, HIF-1α-induced expression and membrane recruitment of the neuronal transporter of glucose GLUT3, and GLUT3-mediated uptake of glucose. These and other data discussed in this Review suggest that the postulated neurotoxic effect of tPA needs to be reconsidered and instead indicate the emergence of a new paradigm: that tPA is an endogenous neuroprotectant in the central nervous system (CNS).
Collapse
Affiliation(s)
- Manuel Yepes
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine and Veterans Affairs Medical Center Atlanta, GA, USA
| |
Collapse
|
23
|
Fredriksson L, Stevenson TK, Su EJ, Ragsdale M, Moore S, Craciun S, Schielke GP, Murphy GG, Lawrence DA. Identification of a neurovascular signaling pathway regulating seizures in mice. Ann Clin Transl Neurol 2015; 2:722-38. [PMID: 26273685 PMCID: PMC4531055 DOI: 10.1002/acn3.209] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/30/2015] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE A growing body of evidence suggests that increased blood-brain barrier (BBB) permeability can contribute to the development of seizures. The protease tissue plasminogen activator (tPA) has been shown to promote BBB permeability and susceptibility to seizures. In this study, we examined the pathway regulated by tPA in seizures. METHODS An experimental model of kainate-induced seizures was used in genetically modified mice, including mice deficient in tPA (tPA (-/-) ), its inhibitor neuroserpin (Nsp (-/-) ), or both (Nsp:tPA (-/-) ), and in mice conditionally deficient in the platelet-derived growth factor receptor alpha (PDGFRα). RESULTS Compared to wild-type (WT) mice, Nsp (-/-) mice have significantly reduced latency to seizure onset and generalization; whereas tPA (-/-) mice have the opposite phenotype, as do Nsp:tPA (-/-) mice. Furthermore, interventions that maintain BBB integrity delay seizure propagation, whereas osmotic disruption of the BBB in seizure-resistant tPA (-/-) mice dramatically reduces the time to seizure onset and accelerates seizure progression. The phenotypic differences in seizure progression between WT, tPA (-/-) , and Nsp (-/-) mice are also observed in electroencephalogram recordings in vivo, but absent in ex vivo electrophysiological recordings where regulation of the BBB is no longer necessary to maintain the extracellular environment. Finally, we demonstrate that these effects on seizure progression are mediated through signaling by PDGFRα on perivascular astrocytes. INTERPRETATION Together, these data identify a specific molecular pathway involving tPA-mediated PDGFRα signaling in perivascular astrocytes that regulates seizure progression through control of the BBB. Inhibition of PDGFRα signaling and maintenance of BBB integrity might therefore offer a novel clinical approach for managing seizures.
Collapse
Affiliation(s)
- Linda Fredriksson
- Division of Vascular Biology, Department of Medical Biochemistry & Biophysics, Karolinska Institutet Stockholm, Sweden ; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Tamara K Stevenson
- Department of Molecular and Integrative Physiology, University of Michigan Medical School Ann Arbor, Michigan
| | - Enming J Su
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Margaret Ragsdale
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Shannon Moore
- Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School Ann Arbor, Michigan
| | - Stefan Craciun
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Gerald P Schielke
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan
| | - Geoffrey G Murphy
- Department of Molecular and Integrative Physiology, University of Michigan Medical School Ann Arbor, Michigan ; Molecular and Behavioral Neuroscience Institute, University of Michigan Medical School Ann Arbor, Michigan
| | - Daniel A Lawrence
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School Ann Arbor, Michigan ; Department of Molecular and Integrative Physiology, University of Michigan Medical School Ann Arbor, Michigan
| |
Collapse
|
24
|
Levy AD, Omar MH, Koleske AJ. Extracellular matrix control of dendritic spine and synapse structure and plasticity in adulthood. Front Neuroanat 2014; 8:116. [PMID: 25368556 PMCID: PMC4202714 DOI: 10.3389/fnana.2014.00116] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022] Open
Abstract
Dendritic spines are the receptive contacts at most excitatory synapses in the central nervous system. Spines are dynamic in the developing brain, changing shape as they mature as well as appearing and disappearing as they make and break connections. Spines become much more stable in adulthood, and spine structure must be actively maintained to support established circuit function. At the same time, adult spines must retain some plasticity so their structure can be modified by activity and experience. As such, the regulation of spine stability and remodeling in the adult animal is critical for normal function, and disruption of these processes is associated with a variety of late onset diseases including schizophrenia and Alzheimer's disease. The extracellular matrix (ECM), composed of a meshwork of proteins and proteoglycans, is a critical regulator of spine and synapse stability and plasticity. While the role of ECM receptors in spine regulation has been extensively studied, considerably less research has focused directly on the role of specific ECM ligands. Here, we review the evidence for a role of several brain ECM ligands and remodeling proteases in the regulation of dendritic spine and synapse formation, plasticity, and stability in adults.
Collapse
Affiliation(s)
- Aaron D Levy
- Interdepartmental Neuroscience Program, Yale University New Haven, CT, USA ; Department of Molecular Biophysics and Biochemistry, Yale University New Haven, CT, USA
| | - Mitchell H Omar
- Interdepartmental Neuroscience Program, Yale University New Haven, CT, USA ; Department of Molecular Biophysics and Biochemistry, Yale University New Haven, CT, USA
| | - Anthony J Koleske
- Interdepartmental Neuroscience Program, Yale University New Haven, CT, USA ; Department of Molecular Biophysics and Biochemistry, Yale University New Haven, CT, USA ; Department of Neurobiology, Yale University New Haven, CT, USA
| |
Collapse
|
25
|
Deguchi K, Liu N, Liu W, Omote Y, Kono S, Yunoki T, Deguchi S, Yamashita T, Ikeda Y, Abe K. Pericyte protection by edaravone after tissue plasminogen activator treatment in rat cerebral ischemia. J Neurosci Res 2014; 92:1509-19. [PMID: 24938625 PMCID: PMC4263311 DOI: 10.1002/jnr.23420] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 04/16/2014] [Accepted: 04/27/2014] [Indexed: 12/31/2022]
Abstract
Pericytes play a pivotal role in contraction, mediating inflammation and regulation of blood flow in the brain. In this study, changes of pericytes in the neurovascular unit (NVU) were examined in relation to the effects of exogenous tissue plasminogen activator (tPA) and a free radical scavenger, edaravone. Immunohistochemistry and Western blot analyses showed that the overlap between platelet-derived growth factor receptor β-positive pericytes and N-acetylglucosamine oligomers (NAGO)-positive endothelial cells increased significantly at 4 days after 90 min of transient middle cerebral artery occlusion (tMCAO). The number of pericytes and the overlap with NAGO decreased with tPA but recovered with edaravone 4 days after tMCAO with proliferation. Thus, tPA treatment damaged pericytes, resulting in the detachment from astrocytes and a decrease in glial cell line-derived neurotrophic factor secretion. However, treatment with edaravone greatly improved tPA-induced damage to pericytes. The present study demonstrates that exogenous tPA strongly damages pericytes and destroys the integrity of the NVU, but edaravone treatment can greatly ameliorate such damage after acute cerebral ischemia in rats.
Collapse
Affiliation(s)
- Kentaro Deguchi
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Oh SB, Byun CJ, Yun JH, Jo DG, Carmeliet P, Koh JY, Lee JY. Tissue plasminogen activator arrests Alzheimer's disease pathogenesis. Neurobiol Aging 2014; 35:511-9. [DOI: 10.1016/j.neurobiolaging.2013.09.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 10/26/2022]
|
27
|
Subhadra B, Schaller K, Seeds NW. Neuroserpin up-regulation in the Alzheimer's disease brain is associated with elevated thyroid hormone receptor-β1 and HuD expression. Neurochem Int 2013; 63:476-81. [PMID: 24036060 PMCID: PMC3902180 DOI: 10.1016/j.neuint.2013.08.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 07/31/2013] [Accepted: 08/05/2013] [Indexed: 12/14/2022]
Abstract
Neuroserpin, the major inhibitor of tissue plasminogen activator (tPA) in brain, has been shown to be up-regulated in Alzheimer's disease (AD). Inhibition of tPA activity leads to reduced brain levels of plasmin, one of the main enzymes responsible for the degradation and clearance of amyloid-beta and its plaques from the brain. Thyroid hormone is one of the few factors known to enhance expression of neuroserpin in neurons. Thyroid hormone acts on neurons by binding to its receptors THR1α and THR1β, which then function in the nucleus to up-regulate the expression of numerous genes including the RNA-binding protein HuD. HuD acts post-transcriptionally to enhance expression of numerous proteins including neuroserpin by stabilizing their mRNAs. A series of Alzheimer's disease brain tissues were compared to age-matched control brains for their expression of neuroserpin, THRβ1 and HuD by western blotting. Alzheimer's disease brain tissues with elevated neuroserpin protein also showed increased expression of THRβ1 and HuD. Pair-wise analyses showed significant correlation p-values between neuroserpin, THRβ1 and HuD levels; suggesting that the up-regulation of neuroserpin in Alzheimer's disease brain may result from an activation of the thyroid hormone response system in these individuals. These findings provide evidence for a potential relationship between thyroid hormone disorders and Alzheimer's disease.
Collapse
Affiliation(s)
- Bobban Subhadra
- Cell Biology & Physiology, University of New Mexico HSC, Albuquerque, NM, USA
| | - Kristin Schaller
- Biochemistry & Molecular Genetics, University of Colorado HSC, Denver, CO, USA
| | - Nicholas W. Seeds
- Cell Biology & Physiology, University of New Mexico HSC, Albuquerque, NM, USA
- Biochemistry & Molecular Genetics, University of Colorado HSC, Denver, CO, USA
| |
Collapse
|
28
|
Cops EJ, Sashindranath M, Daglas M, Short KM, da Fonseca Pereira C, Pang TY, Lijnen RH, Smyth IM, Hannan AJ, Samson AL, Medcalf RL. Tissue-type plasminogen activator is an extracellular mediator of Purkinje cell damage and altered gait. Exp Neurol 2013; 249:8-19. [PMID: 23939410 DOI: 10.1016/j.expneurol.2013.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 07/26/2013] [Accepted: 08/01/2013] [Indexed: 12/20/2022]
Abstract
Purkinje neurons are a sensitive and specialised cell type important for fine motor movement and coordination. Purkinje cell damage manifests as motor incoordination and ataxia - a prominent feature of many human disorders including spinocerebellar ataxia and Huntington's disease. A correlation between Purkinje degeneration and excess cerebellar levels of tissue-type plasminogen activator (tPA) has been observed in multiple genetically-distinct models of ataxia. Here we show that Purkinje loss in a mouse model of Huntington's disease also correlates with a 200% increase in cerebellar tPA activity. That elevated tPA levels arise in a variety of ataxia models suggests that tPA is a common mediator of Purkinje damage. To address the specific contribution of tPA to cerebellar dysfunction we studied the T4 mice line that overexpresses murine tPA in postnatal neurons through the Thy1.2 gene promoter, which directs preferential expression to Purkinje cells within the cerebellum. Here we show that T4 mice develop signs of cerebellar damage within 10 weeks of birth including atrophy of Purkinje cell soma and dendrites, astrogliosis, reduced molecular layer volume and altered gait. In contrast, T4 mice displayed no evidence of microgliosis, nor any changes in interneuron density, nor alteration in the cerebellar granular neuron layer. Thus, excess tPA levels may be sufficient to cause targeted Purkinje cell degeneration and ataxia. We propose that elevated cerebellar tPA levels exert a common pathway of Purkinje cell damage. Therapeutically lowering cerebellar tPA levels may represent a novel means of preserving Purkinje cell integrity and motor coordination across a wide range of neurodegenerative diseases.
Collapse
Affiliation(s)
- Elisa J Cops
- Australian Centre for Blood Diseases, AMREP, Monash University, Melbourne, Victoria 3004, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The plasminogen activation system and the regulation of catecholaminergic function. J Biomed Biotechnol 2012; 2012:721657. [PMID: 23097598 PMCID: PMC3477892 DOI: 10.1155/2012/721657] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 05/17/2012] [Indexed: 11/29/2022] Open
Abstract
The local environment of neurosecretory cells contains the major components of the plasminogen activation system, including the plasminogen activators, tissue plasminogen activator (t-PA) and urokinase-type plasminogen activator (u-PA), as well as binding sites for t-PA, the receptor for u-PA (uPAR), and also the plasminogen activator inhibitor, PAI-1. Furthermore, these cells express specific binding sites for plasminogen, which is available in the circulation and in interstitial fluid. Colocalization of plasminogen and its activators on cell surfaces provides a mechanism for promoting local plasminogen activation. Plasmin is retained on the cell surface where it is protected from its inhibitor, α2-antiplasmin. In neurosecretory cells, localized plasmin activity provides a mechanism for extracellular processing of secreted hormones. Neurotransmitter release from catecholaminergic cells is negatively regulated by cleavage products formed by plasmin-mediated proteolysis. Recently, we have identified a major plasminogen receptor, Plg-RKT. We have found that Plg-RKT is highly expressed in chromaffin cells of the adrenal medulla as well as in other catecholaminergic cells and tissues. Plg-RKT-dependent plasminogen activation plays a key role in regulating catecholaminergic neurosecretory cell function.
Collapse
|
30
|
McEwen BS. Structural plasticity of the adult brain: how animal models help us understand brain changes in depression and systemic disorders related to depression. DIALOGUES IN CLINICAL NEUROSCIENCE 2012. [PMID: 22034132 PMCID: PMC3181799 DOI: 10.31887/dcns.2004.6.2/bmcewen] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The brain interprets experiences and translates them into behavioral and physiological responses. Stressful events are those which are threatening or, at the very least, unexpected and surprising, and the physiological and behavioral responses are intended to promote adaptation via a process called “allostasis. ” Chemical mediators of allostasis include cortisol and adrenalin from the adrenal glands, other hormones, and neurotransmitters, the parasympathetic and sympathetic nervous systems, and cytokines and chemokines from the immune system. Two brain structures, the amygdala and hippocampus, play key roles in interpreting what is stressful and determining appropriate responses. The hippocampus, a key structure for memories of events and contexts, expresses receptors that enable it to respond to glucocorticoid hormones in the blood, it undergoes atrophy in a number of psychiatric disorders; it also responds to stressors with changes in excitability, decreased dendritic branching, and reduction in number of neurons in the dentate gyrus. The amygdala, which is important for “emotional memories, ” becomes hyperactive in posttraumatic stress disorder and depressive illness, in animal models of stress, there is evidence for growth and hypertrophy of nerve cells in the amygdala. Changes in the brain after acute and chronic stressors mirror the pattern seen in the metabolic, cardiovascular, and immune systems, that is, short-term adaptation (allostasis) followed by long-term damage (allostatic load), eg, atherosclerosis, fat deposition obesity, bone demineralization, and impaired immune function. Allostatic load of this kind is seen in major depressive illness and may also be expressed in other chronic anxiety and mood disorders.
Collapse
Affiliation(s)
- Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
31
|
Veeravalli KK, Dasari VR, Rao JS. Regulation of proteases after spinal cord injury. J Neurotrauma 2012; 29:2251-62. [PMID: 22709139 DOI: 10.1089/neu.2012.2460] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury is a major medical problem worldwide. Unfortunately, we still do not have suitable therapeutic agents for the treatment of spinal cord injury and prevention of its devastating consequences. Scientists and physicians are baffled by the challenges of controlling progressive neurodegeneration in spinal cord injury, which has not been healed with any currently-available treatments. Although extensive work has been carried out to better understand the pathophysiology of spinal cord injury, our current understanding of the repair mechanisms of secondary injury processes is still meager. Several investigators reported the crucial role played by various proteases after spinal cord injury. Understanding the beneficial and harmful roles these proteases play after spinal cord injury will allow scientists to plan and design appropriate treatment strategies to improve functional recovery after spinal cord injury. This review will focus on various proteases such as matrix metalloproteinases, cysteine proteases, and serine proteases and their inhibitors in the context of spinal cord injury.
Collapse
Affiliation(s)
- Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61605, USA
| | | | | |
Collapse
|
32
|
Pirchl M, Ullrich C, Sperner-Unterweger B, Humpel C. Homocysteine has anti-inflammatory properties in a hypercholesterolemic rat model in vivo. Mol Cell Neurosci 2012; 49:456-63. [PMID: 22425561 PMCID: PMC3359503 DOI: 10.1016/j.mcn.2012.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 01/13/2023] Open
Abstract
Inflammation is a hallmark in many neurodegenerative diseases like Alzheimer's disease or vascular dementia. Cholesterol and homocysteine are both vascular risk factors which have been associated with dementia, inflammation and blood–brain barrier dysfunction. In previous studies we found that hypercholesterolemia but not hyperhomocysteinemia induced inflammation in rats in vivo. The aim of the present study was to investigate the effect of a combined treatment of Sprague Dawley rats with cholesterol and homocysteine for 5 months on spatial learning and memory, blood–brain barrier integrity and inflammation. Cholesterol treated rats showed severe learning deficits, while rats treated with cholesterol and homocysteine (Mix) counteracted the cholesterol-induced inflammation and partly the cortical blood–brain barrier disruptions, although cognition was still impaired. To study the potential protective effect of homocysteine, inflammation was induced in organotypic rat brain cortex slices and primary microglial cells by treatment with different inflammatory stimuli (e.g. lipopolysaccharide or tissue plasminogen activator). Tissue plasminogen activator-induced inflammation was counteracted by homocysteine. In conclusion, our data demonstrate that homocysteine significantly ameliorates cholesterol-induced inflammation and blood–brain barrier disruption but not the memory impairment, possibly involving a tissue plasminogen activator-related mechanism.
Collapse
Affiliation(s)
| | | | | | - Christian Humpel
- Corresponding author at: Dep. of Psychiatry and Psychotherapy, Anichstr. 35, A-6020 Innsbruck, Austria. Fax: + 43 512 504 23713.
| |
Collapse
|
33
|
Modifying neurorepair and neuroregenerative factors with tPA and edaravone after transient middle cerebral artery occlusion in rat brain. Brain Res 2011; 1436:168-77. [PMID: 22221736 DOI: 10.1016/j.brainres.2011.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 12/05/2011] [Accepted: 12/09/2011] [Indexed: 11/20/2022]
Abstract
Changes in expression of neurorepair and neuroregenerative factors were examined after transient cerebral ischemia in relation to the effects of tissue plasminogen activator (tPA) and the free radical scavenger edaravone. Physiological saline or edaravone was injected twice during 90 min of transient middle cerebral artery occlusion (tMCAO) in rats, followed by the same saline or tPA at reperfusion. Sizes of the infarct and protein factors relating to neurorepair and neuroregeneration were examined at 4d after tMCAO. The protein factors examined were: a chondroitin sulfate proteoglycan neurocan, semaphorin type 3A (Sema3A), a myelin-associated glycoprotein receptor (Nogo receptor, Nogo-R), a synaptic regenerative factor (growth associated protein-43, GAP43), and a chemotropic factor netrin receptor (deleted in colorectal cancer, DCC). Two groups treated by edaravone only or edaravone plus tPA showed a reduction in infarct volume compared to the two groups treated by vehicle only or vehicle plus tPA. Immunohistochemistry and western blot analyses indicated that protein expression of neurocan, Sema3A, Nogo-R, GAP43, and DCC was decreased with tPA, but recovered with edaravone. Additive edaravone prevented the reductions of these five proteins induced by tPA. The present study demonstrates for the first time that exogenous tPA reduced protein factors involved in inhibiting and promoting axonal growth, but that edaravone ameliorated such damage in brain repair after acute ischemia.
Collapse
|
34
|
Fabbro S, Schaller K, Seeds NW. Amyloid-beta levels are significantly reduced and spatial memory defects are rescued in a novel neuroserpin-deficient Alzheimer's disease transgenic mouse model. J Neurochem 2011; 118:928-38. [PMID: 21689108 DOI: 10.1111/j.1471-4159.2011.07359.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Amyloid-beta (Aβ) plaques are a hallmark of Alzheimer's disease. Several proteases including plasmin are thought to promote proteolytic cleavage and clearance of Aβ from brain. The activity of both plasmin and tissue plasminogen activator are reduced in Alzheimer's disease brain, while the tissue plasminogen activator inhibitor neuroserpin is up-regulated. Here, the relationship of tissue plasminogen activator and neuroserpin to Aβ levels is explored in mouse models. Aβ(1-42) peptide injected into the frontal cortex of tissue plasminogen activator knockout mice is slow to disappear compared to wildtype mice, whereas neuroserpin knockout mice show a rapid clearance of Aβ(1-42). The relationship of neuroserpin and tissue plasminogen activator to Aβ plaque formation was studied further by knocking-out neuroserpin in the human amyloid precursor protein-J20 transgenic mouse. Compared to the J20-transgenic mouse, the neuroserpin-deficient J20-transgenic mice have a dramatic reduction of Aβ peptides, fewer and smaller plaques, and more active tissue plasminogen activator associated with plaques. Furthermore, neuroserpin-deficient J20-transgenic mice have near normal performances in the Morris water maze, in contrast to the spatial memory defects seen in J20-transgenic mice. These results support the concept that neuroserpin inhibition of tissue plasminogen activator plays an important role both in the accumulation of brain amyloid plaques and loss of cognitive abilities.
Collapse
Affiliation(s)
- Shay Fabbro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | | | | |
Collapse
|
35
|
Obiang P, Maubert E, Bardou I, Nicole O, Launay S, Bezin L, Vivien D, Agin V. Enriched housing reverses age-associated impairment of cognitive functions and tPA-dependent maturation of BDNF. Neurobiol Learn Mem 2011; 96:121-9. [DOI: 10.1016/j.nlm.2011.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 02/22/2011] [Accepted: 03/21/2011] [Indexed: 01/19/2023]
|
36
|
Hong I, Kim J, Song B, Park S, Lee J, Kim J, An B, Lee S, Choi S. Modulation of fear memory by retrieval and extinction: a clue for memory deconsolidation. Rev Neurosci 2011; 22:205-29. [PMID: 21476941 DOI: 10.1515/rns.2011.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Memories are fragile and easily forgotten at first, but after a consolidation period of hours to weeks, are inscribed in our brains as stable traces, no longer vulnerable to conventional amnesic treatments. Retrieval of a memory renders it labile, akin to the early stages of consolidation. This phenomenon has been explored as memory reactivation, in the sense that the memory is temporarily 'deconsolidated', allowing a short time window for amnesic intervention. This window closes again after reconsolidation, which restores the stability of the memory. In contrast to this 'transient deconsolidation' and the short-spanned amnesic effects of consolidation blockers, some specific treatments can disrupt even consolidated memory, leading to apparent amnesia. We propose the term 'amnesic deconsolidation' to describe such processes that lead to disruption of consolidated memory and/or consolidated memory traces. We review studies of these 'amnesic deconsolidation' treatments that enhance memory extinction, alleviate relapse, and reverse learning-induced plasticity. The transient deconsolidation that memory retrieval induces and the amnesic deconsolidation that these regimes induce both seem to dislodge a component that stabilizes consolidated memory. Characterizing this component, at both molecular and network levels, will provide a key to developing clinical treatments for memory-related disorders and to defining the consolidated memory trace.
Collapse
Affiliation(s)
- Ingie Hong
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Bai H, Baik N, Kiosses WB, Krajewski S, Miles LA, Parmer RJ. The novel plasminogen receptor, plasminogen receptor(KT) (Plg-R(KT)), regulates catecholamine release. J Biol Chem 2011; 286:33125-33. [PMID: 21795689 DOI: 10.1074/jbc.m111.218693] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neurotransmitter release by catecholaminergic cells is negatively regulated by prohormone cleavage products formed from plasmin-mediated proteolysis. Here, we investigated the expression and subcellular localization of Plg-R(KT), a novel plasminogen receptor, and its role in catecholaminergic cell plasminogen activation and regulation of catecholamine release. Prominent staining with anti-Plg-R(KT) mAb was observed in adrenal medullary chromaffin cells in murine and human tissue. In Western blotting, Plg-R(KT) was highly expressed in bovine adrenomedullary chromaffin cells, human pheochromocytoma tissue, PC12 pheochromocytoma cells, and murine hippocampus. Expression of Plg-R(KT) fused in-frame to GFP resulted in targeting of the GFP signal to the cell membrane. Phase partitioning, co-immunoprecipitation with urokinase-type plasminogen activator receptor (uPAR), and FACS analysis with antibody directed against the C terminus of Plg-R(KT) were consistent with Plg-R(KT) being an integral plasma membrane protein on the surface of catecholaminergic cells. Cells stably overexpressing Plg-R(KT) exhibited substantial enhancement of plasminogen activation, and antibody blockade of non-transfected PC12 cells suppressed plasminogen activation. In functional secretion assays, nicotine-evoked [(3)H]norepinephrine release from cells overexpressing Plg-R(KT) was markedly decreased (by 51 ± 2%, p < 0.001) when compared with control transfected cells, and antibody blockade increased [(3)H]norepinephrine release from non-transfected PC12 cells. In summary, Plg-R(KT) is present on the surface of catecholaminergic cells and functions to stimulate plasminogen activation and modulate catecholamine release. Plg-R(KT) thus represents a new mechanism and novel control point for regulating the interface between plasminogen activation and neurosecretory cell function.
Collapse
Affiliation(s)
- Hongdong Bai
- Department of Medicine, University of California, San Diego, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
38
|
Sashindranath M, Samson AL, Downes CE, Crack PJ, Lawrence AJ, Li QX, Ng AQP, Jones NC, Farrugia JJ, Abdella E, Vassalli JD, Madani R, Medcalf RL. Compartment- and context-specific changes in tissue-type plasminogen activator (tPA) activity following brain injury and pharmacological stimulation. J Transl Med 2011; 91:1079-91. [PMID: 21519332 DOI: 10.1038/labinvest.2011.67] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Tissue-type plasminogen activator (tPA) is a major protease of the central nervous system. Most studies to date have used in situ- or gel-based zymographic assays to monitor in vivo changes in neural tPA activity. In this study, we demonstrate that the amidolytic assay can be adapted to accurately detect changes in net tPA activity in mouse brain tissues. Using the amidolytic assay, we examined differences in net tPA activity in the cerebral cortex, sub-cortical structures and cerebellum in wildtype (WT) and tPA(-/-) mice, and in transgenic mice selectively overexpressing tPA in neurons. In addition, we assessed changes in endogenous net tPA activity in WT mice following morphine administration, epileptic seizures, traumatic brain injury and ischaemic stroke-neurological settings in which tPA has a known functional role. Under these conditions, acute and compartment-specific regulation of tPA activity was observed. tPA also participates in various forms of chronic neurodegeneration. Accordingly, we assessed tPA activity levels in mouse models of Alzheimer's disease (AD) and spinocerebellar ataxia type-1 (SCA1). Decreased tPA activity was detected in the cortex and subcortex of AD mice, whereas increased tPA activity was found in the cerebellum of SCA1 mice. These findings extend the existing hypotheses that low tPA activity promotes AD, whereas increased tPA activity contributes to cerebellar degeneration. Collectively, our results exemplify the utility of the amidolytic assay and emphasise tPA as a complex mediator of brain function and dysfunction. On the basis of this evidence, we propose that alterations in tPA activity levels could be used as a biomarker for perturbations in brain homeostasis.
Collapse
|
39
|
Gebbink MFBG. Tissue-type plasminogen activator-mediated plasminogen activation and contact activation, implications in and beyond haemostasis. J Thromb Haemost 2011; 9 Suppl 1:174-81. [PMID: 21781253 DOI: 10.1111/j.1538-7836.2011.04278.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Due to its discovery as initiator of fibrinolysis and its well-studied activation by fibrin, tissue-type plasminogen activator (tPA) and the fibrinolytic system are generally associated with the dissolution of blood clots. However, it has been demonstrated over the years that (i) tPA can be activated by multiple proteins, (ii) plasmin has many substrates other than fibrin and (iii) tPA and plasmin have biological functions independent of fibrin and distinct from their role in blood clot lysis. We here review the data with respect to the activation of tPA by fibrin and its multiple other cofactors, in relation to tPA's role in pathophysiology, notably fibrinolysis and amyloidosis, with emphasis on Alzheimer's disease. We demonstrate a common structural element, termed cross-β structure, in misfolded proteins that is causal to tPA activation. The implications for protein misfolding diseases that are known to be associated with the deposition of amyloid and for diseases for which this has not (yet) been established are discussed.
Collapse
Affiliation(s)
- M F B G Gebbink
- Crossbeta Biosciences BV, Utrecht Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
40
|
Almonte AG, Sweatt JD. Serine proteases, serine protease inhibitors, and protease-activated receptors: roles in synaptic function and behavior. Brain Res 2011; 1407:107-22. [PMID: 21782155 DOI: 10.1016/j.brainres.2011.06.042] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/03/2011] [Accepted: 06/16/2011] [Indexed: 12/11/2022]
Abstract
Serine proteases, serine protease inhibitors, and protease-activated receptors have been intensively investigated in the periphery and their roles in a wide range of processes-coagulation, inflammation, and digestion, for example-have been well characterized (see Coughlin, 2000; Macfarlane et al., 2001; Molinari et al., 2003; Wang et al., 2008; Di Cera, 2009 for reviews). A growing number of studies demonstrate that these protein systems are widely expressed in many cell types and regions in mammalian brains. Accumulating lines of evidence suggest that the brain has co-opted the activities of these interesting proteins to regulate various processes underlying synaptic activity and behavior. In this review, we discuss emerging roles for serine proteases in the regulation of mechanisms underlying synaptic plasticity and memory formation.
Collapse
Affiliation(s)
- Antoine G Almonte
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
41
|
Plasminogen activator promotes recovery following spinal cord injury. Cell Mol Neurobiol 2011; 31:961-7. [PMID: 21573723 DOI: 10.1007/s10571-011-9701-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/26/2011] [Indexed: 12/21/2022]
Abstract
Plasminogen activators play an important role in synaptic plasticity associated with the crossed phrenic phenomenon (CPP) and recovery of respiratory function after spinal cord injury. A genetic approach using knockout mice lacking various genes in the plasminogen activator/plasmin system has shown that induction of urokinase plasminogen activator (uPA) is required during the first hour after a C2-hemisection for the acquisition of the CPP response. The uPA knockout mice do not show the structural remodeling of phrenic motor neuron synapses characteristic of the CPP response. As shown here uPA acts in a cell signaling manner via binding to its receptor uPAR rather than as a protease, since uPAR knockout mice or knock-in mice possessing a modified uPA that is unable to bind to uPAR both fail to generate a CPP and recover respiratory function. Microarray data and real-time PCR analysis of mRNAs induced in the phrenic motor nucleus after C2-hemisection in C57Bl/6 mice as compared to uPA knockout mice indicate a potential cell signaling cascade downstream possibly involving β-integrin and Src, and other pathways. Identification of these uPA-mediated signaling pathways may provide the opportunity to pharmacologically upregulate the synaptic plasticity necessary for recovery of phrenic motoneuron activity following cervical spinal cord injury.
Collapse
|
42
|
Kitsukawa T, Nagata M, Yanagihara D, Tomioka R, Utsumi H, Kubota Y, Yagi T, Graybiel AM, Yamamori T. A novel instrumented multipeg running wheel system, Step-Wheel, for monitoring and controlling complex sequential stepping in mice. J Neurophysiol 2011; 106:479-87. [PMID: 21525375 DOI: 10.1152/jn.00139.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Motor control is critical in daily life as well as in artistic and athletic performance and thus is the subject of intense interest in neuroscience. Mouse models of movement disorders have proven valuable for many aspects of investigation, but adequate methods for analyzing complex motor control in mouse models have not been fully established. Here, we report the development of a novel running-wheel system that can be used to evoke simple and complex stepping patterns in mice. The stepping patterns are controlled by spatially organized pegs, which serve as footholds that can be arranged in adjustable, ladder-like configurations. The mice run as they drink water from a spout, providing reward, while the wheel turns at a constant speed. The stepping patterns of the mice can thus be controlled not only spatially, but also temporally. A voltage sensor to detect paw touches is attached to each peg, allowing precise registration of footfalls. We show that this device can be used to analyze patterns of complex motor coordination in mice. We further demonstrate that it is possible to measure patterns of neural activity with chronically implanted tetrodes as the mice engage in vigorous running bouts. We suggest that this instrumented multipeg running wheel (which we name the Step-Wheel System) can serve as an important tool in analyzing motor control and motor learning in mice.
Collapse
Affiliation(s)
- Takashi Kitsukawa
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Shen LH, Xin H, Li Y, Zhang RL, Cui Y, Zhang L, Lu M, Zhang ZG, Chopp M. Endogenous tissue plasminogen activator mediates bone marrow stromal cell-induced neurite remodeling after stroke in mice. Stroke 2011; 42:459-64. [PMID: 21212396 DOI: 10.1161/strokeaha.110.593863] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Bone marrow stromal cells (BMSC) decrease neurological deficits in rodents after stroke and concomitantly induce extensive neurite remodeling in the brain, which highly correlates with the improvement of neurological function. We investigated the effects of endogenous tissue plasminogen activator (tPA) on neurite remodeling after BMSC treatment. METHODS Adult C57BL/6 wild-type (WT) mice and tPA knockout (tPA(-/-)) mice were subjected to middle cerebral artery occlusion, followed by an injection of 1×10(6) BMSC (n=18) or phosphate-buffered saline (n=18) into the tail vein 24 hours later. Behavioral tests were performed at 3, 7, and 14 days after middle cerebral artery occlusion. Animals were euthanized at 14 days after stroke. RESULTS The effects of BMSC on functional recovery depended on presence or absence of tPA, even after adjusting for imbalanced stroke severity. BMSC significantly improve functional recovery in WT mice compared to WT controls but show no beneficial effect in the tPA(-/-) mice compared to tPA(-/-) controls. Axonal density and synaptophysin-positive areas along the ischemic boundary zone of the cortex and striatum in WT mice are significantly higher than in the tPA(-/-) mice. BMSC treatment significantly increases tPA protein level and activity only in WT mice. CONCLUSIONS Our results suggest that endogenous tPA promotes BMSC-induced neurite outgrowth and may contribute to functional recovery after stroke.
Collapse
Affiliation(s)
- Li Hong Shen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Horii-Hayashi N, Yoshikawa M, Matsusue Y, Ishizaka S, Nishi M, Wanaka A. Environmental stimulation changes tissue-type plasminogen activator activity in the adult mouse hippocampus. Neurochem Int 2011; 58:1-4. [DOI: 10.1016/j.neuint.2010.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 10/15/2010] [Accepted: 10/18/2010] [Indexed: 11/28/2022]
|
45
|
Wu J, Echeverry R, Guzman J, Yepes M. Neuroserpin protects neurons from ischemia-induced plasmin-mediated cell death independently of tissue-type plasminogen activator inhibition. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2576-84. [PMID: 20864675 PMCID: PMC2966813 DOI: 10.2353/ajpath.2010.100466] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/15/2010] [Indexed: 11/20/2022]
Abstract
The serine proteinase tissue-type plasminogen activator (tPA) and the serine proteinase inhibitor neuroserpin are both expressed in areas of the brain with the highest vulnerability to hypoxia/ischemia. In vitro studies show that neuroserpin inhibits tPA and, to a lesser extent, urokinase-type plasminogen activator and plasmin. Experimental middle cerebral artery occlusion (MCAO) increases tPA activity and neuroserpin expression in ischemic tissue, and genetic deficiency of tPA or either treatment with or overexpression of neuroserpin decreases the volume of the ischemic lesion following MCAO. These findings have led to the hypothesis that neuroserpin's neuroprotection is mediated by inhibition of tPA's alleged neurotoxic effect. Ischemic preconditioning is a natural adaptive process whereby exposure to a sublethal insult induces tolerance against a subsequent lethal ischemic injury. Here we demonstrate that exposure to sublethal hypoxia/ischemia increases the neuroserpin expression in the hippocampal CA1 layer and cerebral cortex, and that neuroserpin induces ischemic tolerance and decreases the volume of the ischemic lesion following MCAO in wild-type and tPA-deficient (tPA-/-) neurons and mice. Plasmin induces neuronal death, and this effect is abrogated by either neuroserpin or the NMDA receptor antagonist MK-801. Neuroserpin also attenuated kainic acid-induced neuronal death. Our data indicate that the neuroprotective effect of neuroserpin is due to inhibition of plasmin-mediated excitotoxin-induced cell death and is independent of neuroserpin's ability to inhibit tPA activity.
Collapse
Affiliation(s)
- Jialing Wu
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael St, Suite 505J, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
46
|
Nagai N, Matsuo O. Roles of fibrinolytic system components in the nervous system. PATHOPHYSIOLOGY 2010; 17:141-7. [DOI: 10.1016/j.pathophys.2009.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 03/10/2009] [Accepted: 03/23/2009] [Indexed: 10/20/2022] Open
|
47
|
Funktion des fibrinolytischen Systems im Nervensystem und intravasale Fibrinolyse. Hamostaseologie 2010. [DOI: 10.1007/978-3-642-01544-1_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
48
|
Benton RL, Maddie MA, Dincman TA, Hagg T, Whittemore SR. Transcriptional activation of endothelial cells by TGFβ coincides with acute microvascular plasticity following focal spinal cord ischaemia/reperfusion injury. ASN Neuro 2009; 1:e00015. [PMID: 19663807 PMCID: PMC2810814 DOI: 10.1042/an20090008] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 08/07/2009] [Accepted: 08/10/2009] [Indexed: 11/17/2022] Open
Abstract
Microvascular dysfunction, loss of vascular support, ischaemia and sub-acute vascular instability in surviving blood vessels contribute to secondary injury following SCI (spinal cord injury). Neither the precise temporal profile of the cellular dynamics of spinal microvasculature nor the potential molecular effectors regulating this plasticity are well understood. TGFβ (transforming growth factor β) isoforms have been shown to be rapidly increased in response to SCI and CNS (central nervous system) ischaemia, but no data exist regarding their contribution to microvascular dysfunction following SCI. To examine these issues, in the present study we used a model of focal spinal cord ischaemia/reperfusion SCI to examine the cellular response(s) of affected microvessels from 30 min to 14 days post-ischaemia. Spinal endothelial cells were isolated from affected tissue and subjected to focused microarray analysis of TGFβ-responsive/related mRNAs 6 and 24 h post-SCI. Immunohistochemical analyses of histopathology show neuronal disruption/loss and astroglial regression from spinal microvessels by 3 h post-ischaemia, with complete dissolution of functional endfeet (loss of aquaporin-4) by 12 h post-ischaemia. Coincident with this microvascular plasticity, results from microarray analyses show 9 out of 22 TGFβ-responsive mRNAs significantly up-regulated by 6 h post-ischaemia. Of these, serpine 1/PAI-1 (plasminogen-activator inhibitor 1) demonstrated the greatest increase (>40-fold). Furthermore, uPA (urokinase-type plasminogen activator), another member of the PAS (plasminogen activator system), was also significantly increased (>7.5-fold). These results, along with other select up-regulated mRNAs, were confirmed biochemically or immunohistochemically. Taken together, these results implicate TGFβ as a potential molecular effector of the anatomical and functional plasticity of microvessels following SCI.
Collapse
Key Words
- endothelin
- insulin-like growth factor binding protein 3 (igfbp-3)
- interleukin-6 (il-6)
- matrix metalloproteinase 9 (mmp-9)
- plasminogen-activator inhibitor 1 (pai-1)
- urokinase-type plasminogen activator (upa)
- aqp-4, aquaporin-4
- bmp, bone morphogenetic protein
- bscb, blood-spinal cord-barrier
- cns, central nervous system
- ec, endothelial cell
- et, endothelin
- gfap, glial fibrillary acidic protein
- huvec, human umbilical vein endothelial cell
- igf, insulin-like growth factor
- igfbp-3, igf-binding protein 3
- il, interleukin
- lea, lycopersicon esculentum agglutinin
- llc, large latent complex
- map2, microtubule-associated protein 2
- mcao, middle cerebral artery occlusion
- mmp, matrix metalloproteinase
- nvu, neurovascular unit
- pa, plasminogen activator
- pai, pa inhibitor
- pas, pa system
- sci, spinal cord injury
- smvec, spinal microvascular ec
- tbs, tris-buffered saline
- tgfβ, transforming growth factor β
- tpa, tissue-type pa
- tsp-1, thrombospondin-1
- upa, urokinase-type pa
- upar, upa receptor
- vegf, vascular endothelial growth factor
Collapse
Affiliation(s)
- Richard L Benton
- daggerKentucky Spinal Cord Injury Research Center, Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| | | | | | | | | |
Collapse
|
49
|
Synaptic plasticity-associated proteases and protease inhibitors in the brain linked to the processing of extracellular matrix and cell adhesion molecules. ACTA ACUST UNITED AC 2009; 4:223-34. [DOI: 10.1017/s1740925x09990172] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Research on the molecular and cellular basis of learning and memory has focused on the mechanisms that underlie the induction and expression of synaptic plasticity. There is increasing evidence that structural changes at the synapse are associated with synaptic plasticity and that extracellular matrix (ECM) components and cell adhesion molecules are associated with these changes. The functions of both groups of molecules can be regulated by proteolysis. In this article we review the roles of selected proteases and protease inhibitors in perisynaptic proteolysis of the ECM and synaptic adhesion proteins and the impact of proteolysis on synaptic modification and cognitive function.
Collapse
|
50
|
Seeds NW, Akison L, Minor K. Role of plasminogen activator in spinal cord remodeling after spinal cord injury. Respir Physiol Neurobiol 2009; 169:141-9. [PMID: 19651246 DOI: 10.1016/j.resp.2009.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 07/24/2009] [Accepted: 07/25/2009] [Indexed: 12/22/2022]
Abstract
Plasminogen activators play an active role in synaptic plasticity associated with the crossed phrenic phenomenon (CPP) and recovery of respiratory function following spinal cord injury. A genetic approach has been used to identify molecular mechanisms underlying this synaptic plasticity. Knockout mice lacking different genes in the plasminogen activator/plasmin system demonstrate that expression of urokinase plasminogen activator (uPA) is required during the critical 1-2h delay period following C2-hemisection for the acquisition of a good CPP response. uPA knockout mice fail to show the structural remodeling of phrenic motorneuron synapses that underlie the CPP response. Potential mechanisms by which uPA may promote phrenic motorneuron synaptic plasticity have been explored. Expression of uPA receptors, uPAR and LRP-1, are both up-regulated in the ipsilateral phrenic motor nucleus (PMN) following C2-hemisection. A comparison of microarray data and real-time PCR analysis of mRNAs induced in the PMN after hemisection indicate potential cell signaling pathways downstream of uPA's interaction with these cell surface receptors in the PMN. Knowledge of these uPA-mediated signaling pathways may identify potential means for the pharmacological activation of the synaptic plasticity required for recovery of phrenic motorneuron activity.
Collapse
Affiliation(s)
- Nicholas W Seeds
- Department of Biochemistry & Molecular Genetics and Neuroscience Program, University of Colorado School of Medicine, Aurora, CO 80045, United States.
| | | | | |
Collapse
|