1
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Hydrogen Sulfide (H 2S- or H 2S n-Polysulfides) in Synaptic Plasticity: Modulation of NMDA Receptors and Neurotransmitter Release in Learning and Memory. Int J Mol Sci 2025; 26:3131. [PMID: 40243915 PMCID: PMC11988931 DOI: 10.3390/ijms26073131] [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: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Hydrogen sulfide (H2S) has emerged as a pivotal gaseous transmitter in the central nervous system, influencing synaptic plasticity, learning, and memory by modulating various molecular pathways. This review examines recent evidence regarding how H2S regulates NMDA receptor function and neurotransmitter release in neuronal circuits. By synthesizing findings from animal and cellular models, we investigate the impacts of enzymatic H2S production and exogenous H2S on excitatory synaptic currents, long-term potentiation, and intracellular calcium signaling. Data suggest that H2S interacts directly with NMDA receptor subunits, altering receptor function and modulating neuronal excitability. Simultaneously, H2S promotes the release of neurotransmitters such as glutamate and GABA, shaping synaptic dynamics and plasticity. Furthermore, reports indicate that disruptions in H2S metabolism contribute to cognitive impairments and neurodegenerative disorders, underscoring the potential therapeutic value of targeting H2S-mediated pathways. Although the precise mechanisms of H2S-induced changes in synaptic strength remain elusive, a growing body of evidence positions H2S as a significant regulator of memory formation processes. This review calls for more rigorous exploration into the molecular underpinnings of H2S in synaptic plasticity, paving the way for novel pharmacological interventions in cognitive dysfunction.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
2
|
Miyata S, Tsuda M, Mitsui S. Overexpression of Motopsin, an Extracellular Serine Protease Related to Intellectual Disability, Promotes Adult Neurogenesis and Neuronal Responsiveness in the Dentate Gyrus. Mol Neurobiol 2024; 61:4929-4948. [PMID: 38153682 DOI: 10.1007/s12035-023-03890-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
Motopsin, a serine protease encoded by PRSS12, is secreted by neuronal cells into the synaptic clefts in an activity-dependent manner, where it induces synaptogenesis by modulating Na+/K+-ATPase activity. In humans, motopsin deficiency leads to severe intellectual disability and, in mice, it disturbs spatial memory and social behavior. In this study, we investigated mice that overexpressed motopsin in the forebrain using the Tet-Off system (DTG-OE mice). The elevated agrin cleavage or the reduced Na+/K+-ATPase activity was not detected. However, motopsin overexpression led to a reduction in spine density in hippocampal CA1 basal dendrites. While motopsin overexpression decreased the ratio of mature mushroom spines in the DG, it increased the ratio of immature thin spines in CA1 apical dendrites. Female DTG-OE mice showed elevated locomotor activity in their home cages. DTG-OE mice showed aberrant behaviors, such as delayed latency to the target hole in the Barnes maze test and prolonged duration of sniffing objects in the novel object recognition test (NOR), although they retained memory comparable to that of TRE-motopsin littermates, which normally express motopsin. After NOR, c-Fos-positive cells increased in the dentate gyrus (DG) of DTG-OE mice compared with that of DTG-SO littermates, in which motopsin overexpression was suppressed by the administration of doxycycline, and TRE-motopsin littermates. Notably, the numbers of doublecortin- and 5-bromo-2'-deoxyuridine-labeled cells significantly increased in the DG of DTG-OE mice, suggesting increased adult neurogenesis. Importantly, our results revealed a new function in addition to modulating neuronal responsiveness and spine morphology in the DG: the regulation of neurogenesis.
Collapse
Affiliation(s)
- Shiori Miyata
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma, 371-8514, Japan
| | - Masayuki Tsuda
- Division of Laboratory Animal Science, Science Research Center, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi, 783-8505, Japan
| | - Shinichi Mitsui
- Department of Rehabilitation Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa, Maebashi, Gunma, 371-8514, Japan.
| |
Collapse
|
3
|
Faissner A. Low-density lipoprotein receptor-related protein-1 (LRP1) in the glial lineage modulates neuronal excitability. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1190240. [PMID: 37383546 PMCID: PMC10293750 DOI: 10.3389/fnetp.2023.1190240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023]
Abstract
The low-density lipoprotein related protein receptor 1 (LRP1), also known as CD91 or α-Macroglobulin-receptor, is a transmembrane receptor that interacts with more than 40 known ligands. It plays an important biological role as receptor of morphogens, extracellular matrix molecules, cytokines, proteases, protease inhibitors and pathogens. In the CNS, it has primarily been studied as a receptor and clearance agent of pathogenic factors such as Aβ-peptide and, lately, Tau protein that is relevant for tissue homeostasis and protection against neurodegenerative processes. Recently, it was found that LRP1 expresses the Lewis-X (Lex) carbohydrate motif and is expressed in the neural stem cell compartment. The removal of Lrp1 from the cortical radial glia compartment generates a strong phenotype with severe motor deficits, seizures and a reduced life span. The present review discusses approaches that have been taken to address the neurodevelopmental significance of LRP1 by creating novel, lineage-specific constitutive or conditional knockout mouse lines. Deficits in the stem cell compartment may be at the root of severe CNS pathologies.
Collapse
|
4
|
Glucocorticoid-Responsive Tissue Plasminogen Activator (tPA) and Its Inhibitor Plasminogen Activator Inhibitor-1 (PAI-1): Relevance in Stress-Related Psychiatric Disorders. Int J Mol Sci 2023; 24:ijms24054496. [PMID: 36901924 PMCID: PMC10003592 DOI: 10.3390/ijms24054496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Stressful events trigger a set of complex biological responses which follow a bell-shaped pattern. Low-stress conditions have been shown to elicit beneficial effects, notably on synaptic plasticity together with an increase in cognitive processes. In contrast, overly intense stress can have deleterious behavioral effects leading to several stress-related pathologies such as anxiety, depression, substance use, obsessive-compulsive and stressor- and trauma-related disorders (e.g., post-traumatic stress disorder or PTSD in the case of traumatic events). Over a number of years, we have demonstrated that in response to stress, glucocorticoid hormones (GCs) in the hippocampus mediate a molecular shift in the balance between the expression of the tissue plasminogen activator (tPA) and its own inhibitor plasminogen activator inhibitor-1 (PAI-1) proteins. Interestingly, a shift in favor of PAI-1 was responsible for PTSD-like memory induction. In this review, after describing the biological system involving GCs, we highlight the key role of tPA/PAI-1 imbalance observed in preclinical and clinical studies associated with the emergence of stress-related pathological conditions. Thus, tPA/PAI-1 protein levels could be predictive biomarkers of the subsequent onset of stress-related disorders, and pharmacological modulation of their activity could be a potential new therapeutic approach for these debilitating conditions.
Collapse
|
5
|
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
|
6
|
Varangot A, Lebatard S, Bellemain-Sagnard M, Lebouvier L, Hommet Y, Vivien D. Modulations of the neuronal trafficking of tissue-type plasminogen activator (tPA) influences glutamate release. Cell Death Dis 2023; 14:34. [PMID: 36650132 PMCID: PMC9845363 DOI: 10.1038/s41419-022-05543-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
The discovery of the neuronal expression of the serine protease tissue-type plasminogen activator (tPA) has opened new avenues of research, with important implications in the physiopathology of the central nervous system. For example, the interaction of tPA with synaptic receptors (NMDAR, LRP1, Annexin II, and EGFR) and its role in the maturation of BDNF have been reported to influence synaptic plasticity and neuronal survival. However, the mechanisms regulating the neuronal trafficking of tPA are unknown. Here, using high-resolution live cell imaging and a panel of innovative genetic approaches, we first unmasked the dynamic characteristics of the dendritic and axonal trafficking of tPA-containing vesicles under different paradigms of neuronal activation or inhibition. We then report a constitutive exocytosis of tPA- and VAMP2-positive vesicles, dramatically increased in conditions of neuronal activation, with a pattern which was mainly dendritic and thus post-synaptic. We also observed that the synaptic release of tPA led to an increase of the exocytosis of VGlut1 positive vesicles containing glutamate. Finally, we described alterations of the trafficking and exocytosis of neuronal tPA in cultured cortical neurons prepared from tau-22 transgenic mice (a preclinical model of Alzheimer's disease (AD)). Altogether, these data provide new insights about the neuronal trafficking of tPA, contributing to a better knowledge of the tPA-dependent brain functions and dysfunctions.
Collapse
Affiliation(s)
- Alexandre Varangot
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Simon Lebatard
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Mathys Bellemain-Sagnard
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France.
- Department of clinical research, Caen-Normandie University Hospital, CHU, Caen, France.
| |
Collapse
|
7
|
Lépine M, Douceau S, Devienne G, Prunotto P, Lenoir S, Regnauld C, Pouettre E, Piquet J, Lebouvier L, Hommet Y, Maubert E, Agin V, Lambolez B, Cauli B, Ali C, Vivien D. Parvalbumin interneuron-derived tissue-type plasminogen activator shapes perineuronal net structure. BMC Biol 2022; 20:218. [PMID: 36199089 PMCID: PMC9535866 DOI: 10.1186/s12915-022-01419-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Perineuronal nets (PNNs) are specialized extracellular matrix structures mainly found around fast-spiking parvalbumin (FS-PV) interneurons. In the adult, their degradation alters FS-PV-driven functions, such as brain plasticity and memory, and altered PNN structures have been found in neurodevelopmental and central nervous system disorders such as Alzheimer’s disease, leading to interest in identifying targets able to modify or participate in PNN metabolism. The serine protease tissue-type plasminogen activator (tPA) plays multifaceted roles in brain pathophysiology. However, its cellular expression profile in the brain remains unclear and a possible role in matrix plasticity through PNN remodeling has never been investigated. Result By combining a GFP reporter approach, immunohistology, electrophysiology, and single-cell RT-PCR, we discovered that cortical FS-PV interneurons are a source of tPA in vivo. We found that mice specifically lacking tPA in FS-PV interneurons display denser PNNs in the somatosensory cortex, suggesting a role for tPA from FS-PV interneurons in PNN remodeling. In vitro analyses in primary cultures of mouse interneurons also showed that tPA converts plasminogen into active plasmin, which in turn, directly degrades aggrecan, a major structural chondroitin sulfate proteoglycan (CSPG) in PNNs. Conclusions We demonstrate that tPA released from FS-PV interneurons in the central nervous system reduces PNN density through CSPG degradation. The discovery of this tPA-dependent PNN remodeling opens interesting insights into the control of brain plasticity. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01419-8.
Collapse
Affiliation(s)
- Matthieu Lépine
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Sara Douceau
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Gabrielle Devienne
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Sophie Lenoir
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Caroline Regnauld
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Elsa Pouettre
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Juliette Piquet
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Eric Maubert
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Véronique Agin
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Bertrand Lambolez
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Bruno Cauli
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France.
| | - Denis Vivien
- Department of clinical research, CHU de Caen Normandie, Caen, France
| |
Collapse
|
8
|
Tomadesso C, de Lizarrondo SM, Ali C, Landeau B, Mézenge F, Perrotin A, de La Sayette V, Vivien D, Chételat G. Plasma Levels of Tissue-Type Plasminogen Activator (tPA) in Normal Aging and Alzheimer's Disease: Links With Cognition, Brain Structure, Brain Function and Amyloid Burden. Front Aging Neurosci 2022; 14:871214. [PMID: 35747448 PMCID: PMC9211060 DOI: 10.3389/fnagi.2022.871214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Tissue-type plasminogen activator (tPA) is a protease known for its fibrinolytic action but is also involved in physiological and pathophysiological aging processes; including amyloid elimination and synaptic plasticity. The aim of the study was to investigate the role of tPA in cognitive and brain aging. Therefore, we assessed the links between tPA plasma concentration and cognition, structural MRI, FDG-PET and Flobetapir-PET neuroimaging in 155 cognitively unimpaired adults (CUA, aged 20-85 years old) and 32 patients with Alzheimer's disease (ALZ). A positive correlation was found between tPA and age in CUA (p < 0.001), with males showing higher tPA than females (p = 0.05). No significant difference was found between ALZ patients and cognitively unimpaired elders (CUE). Plasma tPA in CUA negatively correlated with global brain volume. No correlation was found with brain FDG metabolism or amyloid deposition. Age-related tPA changes were associated to changes in blood pressure, glycemia and body mass index. Within the ALZ patients, tPA didn't correlate with any cognitive or neuroimaging measures, but only with physiological measures. Altogether our study suggests that increased tPA plasma concentration with age is related to neuronal alterations and cardiovascular risk factors.
Collapse
Affiliation(s)
- Clémence Tomadesso
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
- Department of Clinical Research, CHU Caen-Normandie, Caen, France
| | - Sara Martinez de Lizarrondo
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
| | - Brigitte Landeau
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
| | - Florence Mézenge
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
| | - Audrey Perrotin
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
| | - Vincent de La Sayette
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
- Department of Clinical Research, CHU Caen-Normandie, Caen, France
| | - Gaël Chételat
- Normandie Univ, UNICAEN, INSERM, UMR-S U1237, PHIND, Blood and Brain @ Caen Normandy Institute, Caen, France
- *Correspondence: Gaël Chételat
| |
Collapse
|
9
|
PKCδ-positive GABAergic neurons in the central amygdala exhibit tissue-type plasminogen activator: role in the control of anxiety. Mol Psychiatry 2022; 27:2197-2205. [PMID: 35145231 DOI: 10.1038/s41380-022-01455-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 11/08/2022]
Abstract
Tissue plasminogen activator (tPA) is a serine protease expressed in several brain regions and reported to be involved in the control of emotional and cognitive functions. Nevertheless, little is known about the structure-function relationships of these tPA-dependent behaviors. Here, by using a new model of constitutive tPA-deficient mice (tPAnull), we first show that tPA controls locomotor activity, spatial cognition and anxiety. To investigate the brain structures involved in these tPA-dependent behavioral phenotypes, we next generated tPAflox mice allowing conditional tPA deletion (cKO) following stereotaxic injections of adeno-associated virus driving Cre-recombinase expression (AAV-Cre-GFP). We demonstrate that tPA removal in the dentate gyrus of the hippocampus induces hyperactivity and partial spatial memory deficits. Moreover, the deletion of tPA in the central nucleus of the amygdala, but not in the basolateral nucleus, induces hyperactivity and reduced anxiety-like level. Importantly, we prove that these behaviors depend on the tPA present in the adult brain and not on neurodevelopmental disorders. Also, interestingly, our data show that tPA from Protein kinase-C delta-positive (PKCδ) GABAergic interneurons of the lateral/ capsular part of adult mouse central amygdala controls emotional functions through neuronal activation of the medial central amygdala. Together, our study brings new data about the critical central role of tPA in behavioral modulations in adult mice.
Collapse
|
10
|
Stevenson TK, Moore SJ, Murphy GG, Lawrence DA. Tissue Plasminogen Activator in Central Nervous System Physiology and Pathology: From Synaptic Plasticity to Alzheimer's Disease. Semin Thromb Hemost 2021; 48:288-300. [DOI: 10.1055/s-0041-1740265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
AbstractTissue plasminogen activator's (tPA) fibrinolytic function in the vasculature is well-established. This specific role for tPA in the vasculature, however, contrasts with its pleiotropic activities in the central nervous system. Numerous physiological and pathological functions have been attributed to tPA in the central nervous system, including neurite outgrowth and regeneration; synaptic and spine plasticity; neurovascular coupling; neurodegeneration; microglial activation; and blood–brain barrier permeability. In addition, multiple substrates, both plasminogen-dependent and -independent, have been proposed to be responsible for tPA's action(s) in the central nervous system. This review aims to dissect a subset of these different functions and the different molecular mechanisms attributed to tPA in the context of learning and memory. We start from the original research that identified tPA as an immediate-early gene with a putative role in synaptic plasticity to what is currently known about tPA's role in a learning and memory disorder, Alzheimer's disease. We specifically focus on studies demonstrating tPA's involvement in the clearance of amyloid-β and neurovascular coupling. In addition, given that tPA has been shown to regulate blood–brain barrier permeability, which is perturbed in Alzheimer's disease, this review also discusses tPA-mediated vascular dysfunction and possible alternative mechanisms of action for tPA in Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Tamara K. Stevenson
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Shannon J. Moore
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Geoffrey G. Murphy
- Michigan Neuroscience Institute, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel A. Lawrence
- Department of Molecular and Integrative Physiology, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
11
|
Medhat BM, Abu-Zaid MH, Dorgham D, El-Ghobashy N, Afifi AY, El-Makawi S, Ayoub DR, Khalaf OO, Amer R, Koptan DMT, Maged LA. Prevalence of Anti-Nuclear Antibodies and Anti-Phospholipid Antibodies in an Egyptian Cohort with Schizophrenia: A Case-Control Study. Curr Rheumatol Rev 2021; 18:266-271. [PMID: 34751124 DOI: 10.2174/1573397117666211109115120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/19/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Psychiatric disorders, including schizophrenia could herald other manifestation(s) of systemic lupus erythematosus (SLE) potentially hindering timely and optimal management. Moreover, schizophrenia is among the described 'extra-criteria' manifestations of anti-phospholipid syndrome (APS). Hence, screening schizophrenia patients for SLE and APS may pose diagnostic and therapeutic implications. OBJECTIVES Examine schizophrenia patients with no overt connective tissue disease(s) manifestation(s) for clinical and/or serologic evidence of SLE and/or APS. METHODS The study included 92 schizophrenia patients [61 (66.3%) males] and 100 age- and gender-matched healthy controls. Both groups were tested for anti-nuclear antibodies (ANAs), anti-double stranded deoxyribonucleic acid (anti-dsDNA) antibodies, complement 3 (C3) and C4, and criteria anti-phospholipid antibodies (aPL) [anticardiolipin Immunoglobulin (Ig) G and IgM, anti-beta-2-glycoprotein I IgG and IgM, and lupus anticoagulant (LAC)]. RESULTS The patients' mean age and disease duration were 28.8 ± 8.1 and 5.7 ± 2.2 years, respectively. The prevalence of ANA positivity, height of titre, and pattern was comparable between patients and controls (p = 0.9, p = 0.8 and p = 0.1, respectively). Anti-dsDNA antibodies and hypocomplementemia were absent in both groups. A significantly higher frequency of positive LAC was observed among patients compared with controls (7.6 % vs. 1 %, p = 0.02), whereas other aPL were comparable between both groups. None of the patients or controls demonstrated clinically meaningful (medium or high) aPL titres. CONCLUSION In our study, schizophrenia was solely associated with LAC. Thus, in the absence of findings suggestive of SLE or APS, routine screening for both diseases is questionable.
Collapse
Affiliation(s)
- Basma M Medhat
- Rheumatology and Rehabilitation Department, Al Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Mohammed H Abu-Zaid
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Tanta University, Tanta. Egypt
| | - Dalia Dorgham
- Rheumatology and Rehabilitation Department, Al Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Nehal El-Ghobashy
- Rheumatology and Rehabilitation Department, Al Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Angie Yousri Afifi
- Rheumatology and Rehabilitation Department, Al Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Shirin El-Makawi
- Psychiatry Department, Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Doaa R Ayoub
- Psychiatry Department, Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Ola O Khalaf
- Psychiatry Department, Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Reham Amer
- Neuropsychiatry Department, Faculty of Medicine, Tanta University, Tanta. Egypt
| | - Dina M T Koptan
- Clinical and Chemical Pathology Department, Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| | - Lobna A Maged
- Rheumatology and Rehabilitation Department, Al Kasr Alainy Faculty of Medicine, Cairo University, Cairo. Egypt
| |
Collapse
|
12
|
Price R, Mercuri NB, Ledonne A. Emerging Roles of Protease-Activated Receptors (PARs) in the Modulation of Synaptic Transmission and Plasticity. Int J Mol Sci 2021; 22:E869. [PMID: 33467143 PMCID: PMC7830300 DOI: 10.3390/ijms22020869] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/22/2022] Open
Abstract
Protease-activated receptors (PARs) are a class of G protein-coupled receptors (GPCRs) with a unique mechanism of activation, prompted by a proteolytic cleavage in their N-terminal domain that uncovers a tethered ligand, which binds and stimulates the same receptor. PARs subtypes (PAR1-4) have well-documented roles in coagulation, hemostasis, and inflammation, and have been deeply investigated for their function in cellular survival/degeneration, while their roles in the brain in physiological conditions remain less appreciated. Here, we describe PARs' effects in the modulation of neurotransmission and synaptic plasticity. Available evidence, mainly concerning PAR1-mediated and PAR2-mediated regulation of glutamatergic and GABAergic transmission, supports that PARs are important modulators of synaptic efficacy and plasticity in normal conditions.
Collapse
Affiliation(s)
- Rachel Price
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy; (R.P.); (N.B.M.)
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy; (R.P.); (N.B.M.)
- Department of Systems Medicine, Università di Roma “Tor Vergata”, 00133 Rome, Italy
| | - Ada Ledonne
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, 00143 Rome, Italy; (R.P.); (N.B.M.)
| |
Collapse
|
13
|
Baker SK, Strickland S. A critical role for plasminogen in inflammation. J Exp Med 2020; 217:133866. [PMID: 32159743 PMCID: PMC7144526 DOI: 10.1084/jem.20191865] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/10/2019] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Plasminogen and its active form, plasmin, have diverse functions related to the inflammatory response in mammals. Due to these roles in inflammation, plasminogen has been implicated in the progression of a wide range of diseases with an inflammatory component. In this review, we discuss the functions of plasminogen in inflammatory regulation and how this system plays a role in the pathogenesis of diseases spanning organ systems throughout the body.
Collapse
Affiliation(s)
- Sarah K Baker
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY
| |
Collapse
|
14
|
Pasquet N, Douceau S, Naveau M, Lesept F, Louessard M, Lebouvier L, Hommet Y, Vivien D, Bardou I. Tissue-Type Plasminogen Activator Controlled Corticogenesis Through a Mechanism Dependent of NMDA Receptors Expressed on Radial Glial Cells. Cereb Cortex 2020; 29:2482-2498. [PMID: 29878094 DOI: 10.1093/cercor/bhy119] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Indexed: 01/24/2023] Open
Abstract
Modifications of neuronal migration during development, including processes that control cortical lamination are associated with functional deficits at adult stage. Here, we report for the first time that the lack of the serine protease tissue-type Plasminogen Activator (tPA), previously characterized as a neuromodulator and a gliotransmitter, leads to an altered cortical lamination in adult. This results in a neuronal migration defect of tPA deficient neurons which are stopped in the intermediate zone at E16. This phenotype is rescued by re-expressing a wild-type tPA in cortical neurons at E14 but not by a tPA that cannot interact with NMDAR. We thus hypothetized that the tPA produced by cortical neuronal progenitors can control their own radial migration through a mechanism dependent of NMDAR expressed at the surface of radial glial cells (RGC). Accordingly, conditional deletion of tPA in neuronal progenitors at E14 or overexpression of a dominant-negative NMDAR that cannot bind tPA in RGC also delayed neuronal migration. Moreover, the lack of tPA lead to an impaired maturation and orientation of RGC. These data provide the first demonstration that the neuronal serine protease tPA is an actor of a proper corticogenesis by its ability to control NMDAR signaling in RGC.
Collapse
Affiliation(s)
- Nolwenn Pasquet
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Sara Douceau
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Mickael Naveau
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Flavie Lesept
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Morgane Louessard
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Laurent Lebouvier
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Yannick Hommet
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| | - Denis Vivien
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France.,CHU Caen, Clinical Research Department, Caen University Hospital, Caen, France
| | - Isabelle Bardou
- Normandie Université, UNICAEN, INSERM, INSERM UMR-S U 1237, "Physiopathology and Imaging of Neurological Disorders", GIP Cyceron, Caen, France
| |
Collapse
|
15
|
Price R, Ferrari E, Gardoni F, Mercuri NB, Ledonne A. Protease-activated receptor 1 (PAR1) inhibits synaptic NMDARs in mouse nigral dopaminergic neurons. Pharmacol Res 2020; 160:105185. [PMID: 32891865 DOI: 10.1016/j.phrs.2020.105185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022]
Abstract
Protease-activated receptor 1 (PAR1) is a G protein-coupled receptor (GPCR), whose activation requires a proteolytic cleavage in the extracellular domain exposing a tethered ligand, which binds to the same receptor thus stimulating Gαq/11-, Gαi/o- and Gα12-13 proteins. PAR1, activated by serine proteases and matrix metalloproteases, plays multifaceted roles in neuroinflammation and neurodegeneration, in stroke, brain trauma, Alzheimer's diseases, and Parkinson's disease (PD). Substantia nigra pars compacta (SNpc) is among areas with highest PAR1 expression, but current evidence on its roles herein is restricted to mechanisms controlling dopaminergic (DAergic) neurons survival, with controversial data showing PAR1 either fostering or counteracting degeneration in PD models. Since PAR1 functions on SNpc DAergic neurons activity are unknown, we investigated if PAR1 affects glutamatergic transmission in this neuronal population. We analyzed PAR1's effects on NMDARs and AMPARs by patch-clamp recordings from DAergic neurons from mouse midbrain slices. Then, we explored subunit composition of PAR1-sensitive NMDARs, with selective antagonists, and mechanisms underlying PAR1-induced NMDARs modulation, by quantifying NMDARs surface expression. PAR1 activation inhibits synaptic NMDARs in SNpc DAergic neurons, without affecting AMPARs. PAR1-sensitive NMDARs contain GluN2B/GluN2D subunits. Moreover, PAR1-mediated NMDARs hypofunction is reliant on NMDARs internalization, as PAR1 stimulation increases NMDARs intracellular levels and pharmacological limitation of NMDARs endocytosis prevents PAR1-induced NMDARs inhibition. We reveal that PAR1 regulates glutamatergic transmission in midbrain DAergic cells. This might have implications in brain's DA-dependent functions and in neurological/psychiatric diseases linked to DAergic dysfunctions.
Collapse
Affiliation(s)
- Rachel Price
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, Università di Roma Tor Vergata, Rome, Italy
| | - Elena Ferrari
- Department of Pharmacological and Biomolecolar Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecolar Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy; Department of Systems Medicine, Università di Roma Tor Vergata, Rome, Italy
| | - Ada Ledonne
- Department of Experimental Neuroscience, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
16
|
Shmakova AA, Rubina KA, Rysenkova KD, Gruzdeva AM, Ivashkina OI, Anokhin KV, Tkachuk VA, Semina EV. Urokinase receptor and tissue plasminogen activator as immediate-early genes in pentylenetetrazole-induced seizures in the mouse brain. Eur J Neurosci 2019; 51:1559-1572. [PMID: 31587391 DOI: 10.1111/ejn.14584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/13/2019] [Accepted: 09/20/2019] [Indexed: 11/30/2022]
Abstract
Epileptogenesis progressively leads to the rearrangement of normal neuronal networks into more excitable ones and can be viewed as a form of neuroplasticity, the molecular mechanisms of which still remain obscure. Here, we studied pentylenetetrazole seizure-induced regulation of genes for plasminogen activator system in the mouse brain. We found that expression of tissue plasminogen activator (tPA) and urokinase receptor (uPAR) mRNA was strongly increased in the mouse cerebral cortex, hippocampus, striatum and amygdala as early as 3 hr after pentylenetetrazole seizures. Such early activity-induced expression of uPAR in the central nervous system has not been demonstrated before. uPAR mRNA accumulation was followed by elevation of uPAR protein, indicating a complete transcription-translation process. Both tPA gene induction and uPAR gene induction were independent of the protein synthesis, suggesting that they are regulated by neural activity as immediate-early genes. In contrast to tPA and uPAR genes, the expression of which returned to the basal level 6 hr following seizures, urokinase and plasminogen activator inhibitor-1 gene expression showed a delayed activation only at 3 days after seizures. In conclusion, our results suggest an important sensitivity of the brain plasminogen activator system to seizure activity which raises the question of its role in activity-dependent neural tissue remodeling in pathological and normal conditions.
Collapse
Affiliation(s)
- Anna A Shmakova
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Kseniya A Rubina
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Karina D Rysenkova
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Anna M Gruzdeva
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russian Federation.,Laboratory for Neurobiology of Memory, P.K. Anokhin Research Institute of Normal Physiology, Moscow, Russian Federation
| | - Olga I Ivashkina
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russian Federation.,Laboratory for Neurobiology of Memory, P.K. Anokhin Research Institute of Normal Physiology, Moscow, Russian Federation
| | - Konstantin V Anokhin
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russian Federation.,Laboratory for Neurobiology of Memory, P.K. Anokhin Research Institute of Normal Physiology, Moscow, Russian Federation
| | - Vsevolod A Tkachuk
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation.,Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Ekaterina V Semina
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation.,Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russian Federation
| |
Collapse
|
17
|
Krishnaswamy VR, Benbenishty A, Blinder P, Sagi I. Demystifying the extracellular matrix and its proteolytic remodeling in the brain: structural and functional insights. Cell Mol Life Sci 2019; 76:3229-3248. [PMID: 31197404 PMCID: PMC11105229 DOI: 10.1007/s00018-019-03182-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022]
Abstract
The extracellular matrix (ECM) plays diverse roles in several physiological and pathological conditions. In the brain, the ECM is unique both in its composition and in functions. Furthermore, almost all the cells in the central nervous system contribute to different aspects of this intricate structure. Brain ECM, enriched with proteoglycans and other small proteins, aggregate into distinct structures around neurons and oligodendrocytes. These special structures have cardinal functions in the normal functioning of the brain, such as learning, memory, and synapse regulation. In this review, we have compiled the current knowledge about the structure and function of important ECM molecules in the brain and their proteolytic remodeling by matrix metalloproteinases and other enzymes, highlighting the special structures they form. In particular, the proteoglycans in brain ECM, which are essential for several vital functions, are emphasized in detail.
Collapse
Affiliation(s)
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Blinder
- Neurobiology, Biochemistry and Biophysics School, Tel Aviv University, Tel Aviv, Israel
- Sagol School for Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
18
|
Lenoir S, Varangot A, Lebouvier L, Galli T, Hommet Y, Vivien D. Post-synaptic Release of the Neuronal Tissue-Type Plasminogen Activator (tPA). Front Cell Neurosci 2019; 13:164. [PMID: 31105531 PMCID: PMC6491899 DOI: 10.3389/fncel.2019.00164] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/08/2019] [Indexed: 11/13/2022] Open
Abstract
The neuronal serine protease tissue-type Plasminogen Activator (tPA) is an important player of the neuronal survival and of the synaptic plasticity. Thus, a better understanding the mechanisms regulating the neuronal trafficking of tPA is required to further understand how tPA can influence brain functions. Using confocal imaging including living cells and high-resolution cell imaging combined with an innovating labeling of tPA, we demonstrate that the neuronal tPA is contained in endosomal vesicles positives for Rabs and in exosomal vesicles positives for synaptobrevin-2 (VAMP2) in dendrites and axons. tPA-containing vesicles differ in their dynamics with the dendritic tPA containing-vesicles less mobile than the axonal tPA-containing vesicles, these laters displaying mainly a retrograde trafficking. Interestingly spontaneous exocytosis of tPA containing-vesicles occurs largely in dendrites.
Collapse
Affiliation(s)
- Sophie Lenoir
- Physiopathology and Imaging of Neurological Disorders, UNICAEN, INSERM, UMR-S U1237, Normandie Université, Caen, France
| | - Alexandre Varangot
- Physiopathology and Imaging of Neurological Disorders, UNICAEN, INSERM, UMR-S U1237, Normandie Université, Caen, France
| | - Laurent Lebouvier
- Physiopathology and Imaging of Neurological Disorders, UNICAEN, INSERM, UMR-S U1237, Normandie Université, Caen, France
| | - Thierry Galli
- Membrane Traffic in Healthy & Diseased Brain, Center of Psychiatry and Neurosciences, INSERM U894, Sorbonne Paris-Cité, Université Paris Descartes, Paris, France
| | - Yannick Hommet
- Physiopathology and Imaging of Neurological Disorders, UNICAEN, INSERM, UMR-S U1237, Normandie Université, Caen, France
| | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders, UNICAEN, INSERM, UMR-S U1237, Normandie Université, Caen, France.,Department of Clinical Research, Caen University Hospital, CHU Caen, Caen, France
| |
Collapse
|
19
|
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
|
20
|
Bres EE, Faissner A. Low Density Receptor-Related Protein 1 Interactions With the Extracellular Matrix: More Than Meets the Eye. Front Cell Dev Biol 2019; 7:31. [PMID: 30931303 PMCID: PMC6428713 DOI: 10.3389/fcell.2019.00031] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/25/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a biological substrate composed of collagens, proteoglycans and glycoproteins that ensures proper cell migration and adhesion and keeps the cell architecture intact. The regulation of the ECM composition is a vital process strictly controlled by, among others, proteases, growth factors and adhesion receptors. As it appears, ECM remodeling is also essential for proper neuronal and glial development and the establishment of adequate synaptic signaling. Hence, disturbances in ECM functioning are often present in neurodegenerative diseases like Alzheimer’s disease. Moreover, mutations in ECM molecules are found in some forms of epilepsy and malfunctioning of ECM-related genes and pathways can be seen in, for example, cancer or ischemic injury. Low density lipoprotein receptor-related protein 1 (Lrp1) is a member of the low density lipoprotein receptor family. Lrp1 is involved not only in ligand uptake, receptor mediated endocytosis and lipoprotein transport—functions shared by low density lipoprotein receptor family members—but also regulates cell surface protease activity, controls cellular entry and binding of toxins and viruses, protects against atherosclerosis and acts on many cell signaling pathways. Given the plethora of functions, it is not surprising that Lrp1 also impacts the ECM and is involved in its remodeling. This review focuses on the role of Lrp1 and some of its major ligands on ECM function. Specifically, interactions with two Lrp1 ligands, integrins and tissue plasminogen activator are described in more detail.
Collapse
Affiliation(s)
- Ewa E Bres
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
21
|
Angelucci F, Čechová K, Průša R, Hort J. Amyloid beta soluble forms and plasminogen activation system in Alzheimer's disease: Consequences on extracellular maturation of brain-derived neurotrophic factor and therapeutic implications. CNS Neurosci Ther 2018; 25:303-313. [PMID: 30403004 PMCID: PMC6488905 DOI: 10.1111/cns.13082] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/18/2022] Open
Abstract
Soluble oligomeric forms of amyloid beta (Aβ) play an important role in causing the cognitive deficits in Alzheimer’s disease (AD) by targeting and disrupting synaptic pathways. Thus, the present research is directed toward identifying the neuronal pathways targeted by soluble forms and, accordingly, develops alternative therapeutic strategies. The neurotrophin brain‐derived neurotrophic factor (BDNF) is synthesized as a precursor (pro‐BDNF) which is cleaved extracellularly by plasmin to release the mature form. The conversion from pro‐BDNF to BDNF is an important process that regulates neuronal activity and memory processes. Plasmin‐dependent maturation of BDNF in the brain is regulated by plasminogen activator inhibitor‐1 (PAI‐1), the natural inhibitor of tissue‐type plasminogen activator (tPA). Therefore, tPA/PAI‐1 system represents an important regulator of extracellular BDNF/pro‐BDNF ratio. In this review, we summarize the data on the components of the plasminogen activation system and on BDNF in AD. Moreover, we will hypothesize a possible pathogenic mechanism caused by soluble Aβ forms based on the effects on tPA/PAI‐1 system and on the consequence of an altered conversion from pro‐BDNF to the mature BDNF in the brain of AD patients. Translation into clinic may include a better characterization of the disease stage and future direction on therapeutic targets.
Collapse
Affiliation(s)
- Francesco Angelucci
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Kateřina Čechová
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Richard Průša
- Department of Medical Chemistry and Clinical Biochemistry, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jakub Hort
- Memory Clinic, Department of Neurology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
22
|
Jeanneret V, Ospina JP, Diaz A, Manrique LG, Merino P, Gutierrez L, Torre E, Wu F, Cheng L, Yepes M. Tissue-type plasminogen activator protects the postsynaptic density in the ischemic brain. J Cereb Blood Flow Metab 2018; 38:1896-1910. [PMID: 29547062 PMCID: PMC6259311 DOI: 10.1177/0271678x18764495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cerebral ischemia causes the presynaptic release of tissue-type plasminogen activator (tPA). The postsynaptic density (PSD) is a postsynaptic structure that provides a matrix where signaling transduction of excitatory synapses takes place. The postsynaptic density protein-95 (PSD-95) is the most abundant scaffolding protein in the postsynaptic density (PSD), where it modulates the postsynaptic response to the presynaptic release of glutamate by regulating the anchoring of glutamate receptors to the PSD. We found that tPA induces the local translation of PSD-95 mRNA and the subsequent recruitment of PSD-95 protein to the PSD, via plasminogen-independent activation of TrkB receptors. Our data show that PSD-95 is removed from the PSD during the early stages of cerebral ischemia, and that this effect is abrogated by either the release of neuronal tPA, or intravenous administration of recombinant tPA (rtPA). We report that the effect of tPA on PSD-95 is associated with inhibition of the phosphorylation and recruitment of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors to the PSD, known to amplify the effect of the excitotoxic injury, and that this is followed by TrkB-mediated protection of dendritic spines from the harmful effects of the hypoxic insult. These data reveal that tPA is a synaptic protector in the ischemic brain.
Collapse
Affiliation(s)
- Valerie Jeanneret
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - Juan P Ospina
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ariel Diaz
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Luis G Manrique
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Paola Merino
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Laura Gutierrez
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA
| | - Enrique Torre
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Fang Wu
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Lihong Cheng
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Manuel Yepes
- 1 Department of Neurology & Center for Neurodegenerative Disease, School of Medicine, Emory University, Atlanta, GA, USA.,2 Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA, USA.,3 Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| |
Collapse
|
23
|
Ito Y, Noguchi K, Morishima Y, Yamaguchi K. Generation and characterization of tissue-type plasminogen activator transgenic rats. J Thromb Thrombolysis 2018; 45:77-87. [PMID: 29168147 PMCID: PMC5756269 DOI: 10.1007/s11239-017-1582-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
To address a species difference in the responsiveness to human recombinant tissue-type plasminogen activator (rt-PA) between rats and humans, tPA transgenic (Tg) rats were generated and characterized. In the rats, transcriptional regulation of tPA was designed under the control of the endogenous tPA promoter. There were no significant differences in hematological parameters between the tPA Tg and non Tg rats. Plasma tPA concentration was significantly increased and serum free PAI-1 was significantly decreased in the tPA Tg rats. Significant overexpression of tPA mRNA in five major organs was also confirmed in the tPA Tg rats. In contrast, the extent of tPA mRNA induction by pathophysiological stimuli (focal cerebral ischemia) was comparable in the two strains. Earlier increase in the plasma D-Dimer level was observed in the tPA Tg rats in a model of thromboembolism compared with the non Tg rats. On the other hand, there was no statistically significant prolongation of bleeding time in a rat model of bleeding between the two strains. rt-PA showed dose-related blood flow restoration in a rat model of thromboembolic stroke in the tPA Tg rats from a dose (1 mg/kg, i.v.) similar to clinical doses for human stroke patients. In conclusion, tPA Tg rats, in which tPA is overexpressed and endogenous fibrinolytic activity is enhanced without hemostatic abnormality, were generated. tPA Tg rats would be beneficial for the pharmacological and the toxicological evaluation of rt-PA and other various fibrinolytic enhancers.
Collapse
Affiliation(s)
- Yusuke Ito
- Rare Disease & LCM Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan.
| | - Kengo Noguchi
- Pharmacovigilance Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Kyoji Yamaguchi
- Rare Disease & LCM Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58 Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| |
Collapse
|
24
|
Hébert M, Anfray A, Chevilley A, Martinez de Lizarrondo S, Quenault A, Louessard M, Roussel BD, Obiang P, Save E, Orset C, Maubert E, Vivien D, Agin V. Distant Space Processing is Controlled by tPA-dependent NMDA Receptor Signaling in the Entorhinal Cortex. Cereb Cortex 2018; 27:4783-4796. [PMID: 27613436 DOI: 10.1093/cercor/bhw275] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/10/2016] [Indexed: 11/14/2022] Open
Abstract
In humans, spatial cognition and navigation impairments are a frequent situation during physiological and pathological aging, leading to a dramatic deterioration in the quality of life. Despite the discovery of neurons with location-specific activity in rodents, that is, place cells in the hippocampus and later on grid cells in the entorhinal cortex (EC), the molecular mechanisms underlying spatial cognition are still poorly known. Our present data bring together in an unusual combination 2 molecules of primary biological importance: a major neuronal excitatory receptor, N-methyl-D-aspartate receptor (NMDAR), and an extracellular protease, tissue plasminogen activator (tPA), in the control of spatial navigation. By using tPA-deficient mice and a structure-selective pharmacological approach, we demonstrate that the tPA-dependent NMDAR signaling potentiation in the EC plays a key and selective role in the encoding and the subsequent use of distant landmarks during spatial learning. We also demonstrate that this novel function of tPA in the EC is reduced during aging. Overall, these results argue for the concept that encoding of proximal versus distal landmarks is mediated not only by different anatomical pathways but also by different molecular mechanisms, with the tPA-dependent potentiation of NMDAR signaling in the EC that plays an important role.
Collapse
Affiliation(s)
- Marie Hébert
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Antoine Anfray
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Arnaud Chevilley
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Sara Martinez de Lizarrondo
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Aurélien Quenault
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Morgane Louessard
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Benoit D Roussel
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Pauline Obiang
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Etienne Save
- Laboratory of Cognitive Neuroscience UMR 7291, Aix-Marseille University, CNRS, 3 Place Victor Hugo, F-13331 Marseille Cedex 3, France
| | - Cyrille Orset
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Eric Maubert
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France.,Biology Department, Clinical Research Department Medical Center, Normandie Université, UNICAEN, 14000 Caen, France
| | - Véronique Agin
- Normandie Univ, UNICAEN, INSERM, Sérine protéases et physiopathologie de l'unité neurovasculaire, 14000 Caen, France
| |
Collapse
|
25
|
Krzisch M, Fülling C, Jabinet L, Armida J, Gebara E, Cassé F, Habbas S, Volterra A, Hornung JP, Toni N. Synaptic Adhesion Molecules Regulate the Integration of New Granule Neurons in the Postnatal Mouse Hippocampus and their Impact on Spatial Memory. Cereb Cortex 2018; 27:4048-4059. [PMID: 27473321 DOI: 10.1093/cercor/bhw217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/21/2016] [Indexed: 11/14/2022] Open
Abstract
Postnatal hippocampal neurogenesis induces network remodeling and may participate to mechanisms of learning. In turn, the maturation and survival of newborn neurons is regulated by their activity. Here, we tested the effect of a cell-autonomous overexpression of synaptic adhesion molecules on the maturation and survival of neurons born postnatally and on hippocampal-dependent memory performances. Families of adhesion molecules are known to induce pre- and post-synaptic assembly. Using viral targeting, we overexpressed three different synaptic adhesion molecules, SynCAM1, Neuroligin-1B and Neuroligin-2A in newborn neurons in the dentate gyrus of 7- to 9-week-old mice. We found that SynCAM1 increased the morphological maturation of dendritic spines and mossy fiber terminals while Neuroligin-1B increased spine density. In contrast, Neuroligin-2A increased both spine density and size as well as GABAergic innervation and resulted in a drastic increase of neuronal survival. Surprisingly, despite increased neurogenesis, mice overexpressing Neuroligin-2A in new neurons showed decreased memory performances in a Morris water maze task. These results indicate that the cell-autonomous overexpression of synaptic adhesion molecules can enhance different aspects of synapse formation on new neurons and increase their survival. Furthermore, they suggest that the mechanisms by which new neurons integrate in the postnatal hippocampus conditions their functional implication in learning and memory.
Collapse
Affiliation(s)
- Marine Krzisch
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Christine Fülling
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Laura Jabinet
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Jan Armida
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Elias Gebara
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Frédéric Cassé
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Samia Habbas
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Andrea Volterra
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Jean-Pierre Hornung
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| | - Nicolas Toni
- Department of Fundamental Neurosciences, University of Lausanne, 9, rue du Bugnon, Lausanne, Switzerland
| |
Collapse
|
26
|
Frühauf-Perez PK, Temp FR, Pillat MM, Signor C, Wendel AL, Ulrich H, Mello CF, Rubin MA. Spermine protects from LPS-induced memory deficit via BDNF and TrkB activation. Neurobiol Learn Mem 2018; 149:135-143. [DOI: 10.1016/j.nlm.2018.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/18/2018] [Accepted: 02/14/2018] [Indexed: 12/18/2022]
|
27
|
Abstract
We all know about classical fibrinolysis, how plasminogen activation by either tissue-type plasminogen activator (t-PA) or urokinase-type plasminogen activator (u-PA) promotes fibrin breakdown, and how this process was harnessed for the therapeutic removal of blood clots. While this is still perfectly true and still applicable to thromboembolic conditions today, another dimension to this system came to light over two decades ago that implicated the plasminogen activating system in a context far removed from the dissolution of blood clots. This unsuspected area related to brain biology where t-PA was linked to a plethora of activities in the CNS, some of which do not necessarily require plasmin generation. Indeed, t-PA either directly or via plasmin, has been shown to not only have key roles in modulating astrocytes, neurons, microglia, and pericytes, but also to have profound effects in a number of CNS conditions, including ischaemic stroke, severe traumatic brain injury and also in neurodegenerative disorders. While compelling insights have been obtained from various animal models, the clinical relevance of aberrant expression of these components in the CNS, although strongly implied, are only just emerging. This review will cover these areas and will also discuss how the use of thrombolytic agents and anti-fibrinolytic drugs may potentially have impacts outside of their clinical intention, particularly in the CNS.
Collapse
Affiliation(s)
- R L Medcalf
- Australian Centre for Blood Diseases, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
28
|
Differences in the Biological Functions of BDNF and proBDNF in the Central Nervous System. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s11055-017-0391-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Krzisch M, Toni N. Forced neuronal interactions cause poor communication. NEUROGENESIS 2017; 4:e1286424. [PMID: 28265586 DOI: 10.1080/23262133.2017.1286424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/13/2017] [Accepted: 01/20/2017] [Indexed: 10/20/2022]
Abstract
Post-natal hippocampal neurogenesis plays a role in hippocampal function, and neurons born post-natally participate to spatial memory and mood control. However, a great proportion of granule neurons generated in the post-natal hippocampus are eliminated during the first 3 weeks of their maturation, a mechanism that depends on their synaptic integration. In a recent study, we examined the possibility of enhancing the synaptic integration of neurons born post-natally, by specifically overexpressing synaptic cell adhesion molecules in these cells. Synaptic cell adhesion molecules are transmembrane proteins mediating the physical connection between pre- and post-synaptic neurons at the synapse, and their overexpression enhances synapse formation. Accordingly, we found that overexpressing synaptic adhesion molecules increased the synaptic integration and survival of newborn neurons. Surprisingly, the synaptic adhesion molecule with the strongest effect on new neurons' survival, Neuroligin-2A, decreased memory performances in a water maze task. We present here hypotheses explaining these surprising results, in the light of the current knowledge of the mechanisms of synaptic integration of new neurons in the post-natal hippocampus.
Collapse
Affiliation(s)
| | - Nicolas Toni
- Department of Fundamental Neuroscience (DNF), University of Lausanne , Lausanne, Switzerland
| |
Collapse
|
30
|
Fredriksson L, Lawrence DA, Medcalf RL. tPA Modulation of the Blood-Brain Barrier: A Unifying Explanation for the Pleiotropic Effects of tPA in the CNS. Semin Thromb Hemost 2017; 43:154-168. [PMID: 27677179 PMCID: PMC5848490 DOI: 10.1055/s-0036-1586229] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The plasminogen activation (PA) system is best known for its role in fibrinolysis. However, it has also been shown to regulate many nonfibrinolytic functions in the central nervous system (CNS). In particular, tissue-type plasminogen activator (tPA) is reported to have pleiotropic activities in the CNS, regulating events such as neuronal plasticity, excitotoxicity, and cerebrovascular barrier integrity, whereas urokinase-type plasminogen activator is mainly associated with tissue remodeling and cell migration. It has been suggested that the role tPA plays in controlling barrier integrity may provide a unifying mechanism for the reported diverse, and often opposing, functions ascribed to tPA in the CNS. Here we will review the possibility that the pleiotropic effects reported for tPA in physiologic and pathologic processes in the CNS may be a consequence of its role in the neurovascular unit in regulation of cerebrovascular responses and subsequently parenchymal homeostasis. We propose that this might offer an explanation for the ongoing debate regarding the neurotoxic versus neuroprotective roles of tPA.
Collapse
Affiliation(s)
- Linda Fredriksson
- Department of Medical Biochemistry & Biophysics, Division of Vascular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Daniel A. Lawrence
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI USA
| | - Robert L. Medcalf
- Molecular Neurotrauma and Haemostasis, Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| |
Collapse
|
31
|
Jeanneret V, Yepes M. Tissue-type plasminogen activator is a homeostatic regulator of synaptic function in the central nervous system. Neural Regen Res 2017; 12:362-365. [PMID: 28469640 PMCID: PMC5399703 DOI: 10.4103/1673-5374.202924] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Membrane depolarization induces the release of the serine proteinase tissue-type plasminogen activator (tPA) from the presynaptic terminal of cerebral cortical neurons. Once in the synaptic cleft this tPA promotes the exocytosis and subsequent endocytic retrieval of glutamate-containing synaptic vesicles, and regulates the postsynaptic response to the presynaptic release of glutamate. Indeed, tPA has a bidirectional effect on the composition of the postsynaptic density (PSD) that does not require plasmin generation or the presynaptic release of glutamate, but varies according to the baseline level of neuronal activity. Hence, in inactive neurons tPA induces phosphorylation and accumulation in the PSD of the Ca2+/calmodulin-dependent protein kinase IIα (pCaMKIIα), followed by pCaMKIIα-induced phosphorylation and synaptic recruitment of GluR1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. In contrast, in active neurons with increased levels of pCaMKIIα in the PSD tPA induces pCaMKIIα and pGluR1 dephosphorylation and their subsequent removal from the PSD. These effects require active synaptic N-methyl-D-aspartate (NMDA) receptors and cyclin-dependent kinase 5 (Cdk5)-induced phosphorylation of the protein phosphatase 1 (PP1) at T320. These data indicate that tPA is a homeostatic regulator of the postsynaptic response of cerebral cortical neurons to the presynaptic release of glutamate via bidirectional regulation of the pCaMKIIα /PP1 switch in the PSD.
Collapse
Affiliation(s)
- Valerie Jeanneret
- Department of Neurology & Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Manuel Yepes
- 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.,Division of Neurosciences, Yerkes National Primate Research Center, Atlanta, GA, USA
| |
Collapse
|
32
|
Lesept F, Chevilley A, Jezequel J, Ladépêche L, Macrez R, Aimable M, Lenoir S, Bertrand T, Rubrecht L, Galea P, Lebouvier L, Petersen KU, Hommet Y, Maubert E, Ali C, Groc L, Vivien D. Tissue-type plasminogen activator controls neuronal death by raising surface dynamics of extrasynaptic NMDA receptors. Cell Death Dis 2016; 7:e2466. [PMID: 27831563 PMCID: PMC5260909 DOI: 10.1038/cddis.2016.279] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/18/2016] [Accepted: 08/02/2016] [Indexed: 11/26/2022]
Abstract
N-methyl-d-aspartate receptors (NMDARs) are ion channels whose synaptic versus extrasynaptic localization critically influences their functions. This distribution of NMDARs is highly dependent on their lateral diffusion at the cell membrane. Each obligatory subunit of NMDARs (GluN1 and GluN2) contains two extracellular clamshell-like domains with an agonist-binding domain and a distal N-terminal domain (NTD). To date, the roles and dynamics of the NTD of the GluN1 subunit in NMDAR allosteric signaling remain poorly understood. Using single nanoparticle tracking in mouse neurons, we demonstrate that the extracellular neuronal protease tissue-type plasminogen activator (tPA), well known to have a role in the synaptic plasticity and neuronal survival, leads to a selective increase of the surface dynamics and subsequent diffusion of extrasynaptic NMDARs. This process explains the previously reported ability of tPA to promote NMDAR-mediated calcium influx. In parallel, we developed a monoclonal antibody capable of specifically blocking the interaction of tPA with the NTD of the GluN1 subunit of NMDAR. Using this original approach, we demonstrate that the tPA binds the NTD of the GluN1 subunit at a lysine in position 178. Accordingly, when applied to mouse neurons, our selected antibody (named Glunomab) leads to a selective reduction of the tPA-mediated surface dynamics of extrasynaptic NMDARs, subsequent signaling and neurotoxicity, both in vitro and in vivo. Altogether, we demonstrate that the tPA is a ligand of the NTD of the obligatory GluN1 subunit of NMDAR acting as a modulator of their dynamic distribution at the neuronal surface and subsequent signaling.
Collapse
Affiliation(s)
- Flavie Lesept
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Arnaud Chevilley
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Julie Jezequel
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux UMR 5297, Bordeaux, France.,CNRS IINS UMR 5297, Bordeaux, France
| | - Laurent Ladépêche
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux UMR 5297, Bordeaux, France.,CNRS IINS UMR 5297, Bordeaux, France
| | - Richard Macrez
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Margaux Aimable
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Sophie Lenoir
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Thomas Bertrand
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | | | | | - Laurent Lebouvier
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | | | - Yannick Hommet
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Eric Maubert
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Carine Ali
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| | - Laurent Groc
- Interdisciplinary Institute for Neuroscience, Université de Bordeaux UMR 5297, Bordeaux, France.,CNRS IINS UMR 5297, Bordeaux, France
| | - Denis Vivien
- Inserm, Inserm UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen-Normandie, GIP Cyceron, Caen, France
| |
Collapse
|
33
|
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
|
34
|
Abbas AK, Villers A, Ris L. Temporal phases of long-term potentiation (LTP): myth or fact? Rev Neurosci 2016; 26:507-46. [PMID: 25992512 DOI: 10.1515/revneuro-2014-0072] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/12/2015] [Indexed: 12/11/2022]
Abstract
Long-term potentiation (LTP) remains the most widely accepted model for learning and memory. In accordance with this belief, the temporal differentiation of LTP into early and late phases is accepted as reflecting the differentiation of short-term and long-term memory. Moreover, during the past 30 years, protein synthesis inhibitors have been used to separate the early, protein synthesis-independent (E-LTP) phase and the late, protein synthesis-dependent (L-LTP) phase. However, the role of these proteins has not been formally identified. Additionally, several reports failed to show an effect of protein synthesis inhibitors on LTP. In this review, a detailed analysis of extensive behavioral and electrophysiological data reveals that the presumed correspondence of LTP temporal phases to memory phases is neither experimentally nor theoretically consistent. Moreover, an overview of the time courses of E-LTP in hippocampal slices reveals a wide variability ranging from <1 h to more than 5 h. The existence of all these conflictual findings should lead to a new vision of LTP. We believe that the E-LTP vs. L-LTP distinction, established with protein synthesis inhibitor studies, reflects a false dichotomy. We suggest that the duration of LTP and its dependency on protein synthesis are related to the availability of a set of proteins at synapses and not to the de novo synthesis of plasticity-related proteins. This availability is determined by protein turnover kinetics, which is regulated by previous and ongoing electrical activities and by energy store availability.
Collapse
|
35
|
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
|
36
|
Şimşek Ş, Çetin İ, Çim A, Kaya S. Elevated levels of tissue plasminogen activator and E-selectin in male children with autism spectrum disorder. Autism Res 2016; 9:1241-1247. [DOI: 10.1002/aur.1638] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/01/2016] [Accepted: 04/04/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Şeref Şimşek
- Department of Child Psychiatry; Dicle University, Medical School; Diyarbakır Turkey
| | - İhsan Çetin
- Department of Nutrition and Dietetics; Batman University, School of Health Sciences; Batman Turkey
| | - Abdullah Çim
- Department of Medical Genetics; Dicle University, Medical School; Diyarbakır Turkey
| | - Savaş Kaya
- Department of Immunology; Dicle University, Medical School; Diyarbakır Turkey
| |
Collapse
|
37
|
Steinberg A, Moreira TP. Neuroendocrinal, Neurodevelopmental, and Embryotoxic Effects of Recombinant Tissue Plasminogen Activator Treatment for Pregnant Women with Acute Ischemic Stroke. Front Neurosci 2016; 10:51. [PMID: 26941596 PMCID: PMC4766278 DOI: 10.3389/fnins.2016.00051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 02/04/2016] [Indexed: 11/23/2022] Open
Abstract
Thrombolysis with recombinant tissue plasminogen activator (rTPA) was the first evidence-based treatment approved for acute stroke. Ischemic stroke is relatively uncommon in fertile women but treatment is often delayed or not given. In randomized trials, pregnancy has been an exclusion criterion for thrombolysis. Physiologic TPA has been shown to have neuroendocrine effects namely in vasopressin secretion. Important TPA effects in brain function and development include neurite outgrowth, migration of cerebellar granular neurons and promotion of long-term potentiation, among others. Until now, no neuroendocrine side-effects have been reported in pregnant women treated with rTPA. The effects of rTPA exposure in the fetus following intravenous thrombolysis in pregnant women are still poorly understood. This depends on low case frequency, short-duration of exposure and the fact that rTPA molecule is too large to pass the placenta. rTPA has a short half-life of 4–5 min, with only 10% of its concentration remaining in circulation after 20 min, which may explain its safety at therapeutically doses. Ischemic stroke during pregnancy occurs most often in the third trimester. Complication rates of rTPA in pregnant women treated for thromboembolic conditions and ischemic stroke were found to be similar when compared to non-pregnant women (7–9% mortality). In embryos of animal models so far, no indications of a teratogenic or mutagenic potential were found. Pregnancy is still considered a relative contraindication when treating acute ischemic stroke with rTPA, however, treatment risk must be balanced against the potential of maternal disability and/or death.
Collapse
Affiliation(s)
- Anna Steinberg
- Department of Neurology, Karolinska University HospitalStockholm, Sweden; Stroke Research Unit, Department of Clinical Neuroscience, Karolinska InstitutetStockholm, Sweden
| | - Tiago P Moreira
- Department of Neurology, Karolinska University HospitalStockholm, Sweden; Stroke Research Unit, Department of Clinical Neuroscience, Karolinska InstitutetStockholm, Sweden
| |
Collapse
|
38
|
In Sickness and in Health: Perineuronal Nets and Synaptic Plasticity in Psychiatric Disorders. Neural Plast 2015; 2016:9847696. [PMID: 26839720 PMCID: PMC4709762 DOI: 10.1155/2016/9847696] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/27/2015] [Indexed: 12/25/2022] Open
Abstract
Rapidly emerging evidence implicates perineuronal nets (PNNs) and extracellular matrix (ECM) molecules that compose or interact with PNNs, in the pathophysiology of several psychiatric disorders. Studies on schizophrenia, autism spectrum disorders, mood disorders, Alzheimer's disease, and epilepsy point to the involvement of ECM molecules such as chondroitin sulfate proteoglycans, Reelin, and matrix metalloproteases, as well as their cell surface receptors. In many of these disorders, PNN abnormalities have also been reported. In the context of the “quadripartite” synapse concept, that is, the functional unit composed of the pre- and postsynaptic terminals, glial processes, and ECM, and of the role that PNNs and ECM molecules play in regulating synaptic functions and plasticity, these findings resonate with one of the most well-replicated aspects of the pathology of psychiatric disorders, that is, synaptic abnormalities. Here we review the evidence for PNN/ECM-related pathology in these disorders, with particular emphasis on schizophrenia, and discuss the hypothesis that such pathology may significantly contribute to synaptic dysfunction.
Collapse
|
39
|
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
|
40
|
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]
|
41
|
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
|
42
|
Robinson SD, Lee TW, Christie DL, Birch NP. Tissue plasminogen activator inhibits NMDA-receptor-mediated increases in calcium levels in cultured hippocampal neurons. Front Cell Neurosci 2015; 9:404. [PMID: 26500501 PMCID: PMC4598481 DOI: 10.3389/fncel.2015.00404] [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: 07/22/2015] [Accepted: 09/23/2015] [Indexed: 01/15/2023] Open
Abstract
NMDA receptors (NMDARs) play a critical role in neurotransmission, acting as essential mediators of many forms of synaptic plasticity, and also modulating aspects of development, synaptic transmission and cell death. NMDAR-induced responses are dependent on a range of factors including subunit composition and receptor location. Tissue-type plasminogen activator (tPA) is a serine protease that has been reported to interact with NMDARs and modulate NMDAR activity. In this study we report that tPA inhibits NMDAR-mediated changes in intracellular calcium levels in cultures of primary hippocampal neurons stimulated by low (5 μM) but not high (50 μM) concentrations of NMDA. tPA also inhibited changes in calcium levels stimulated by presynaptic release of glutamate following treatment with bicucculine/4-aminopyridine (4-AP). Inhibition was dependent on the proteolytic activity of tPA but was unaffected by α2-antiplasmin, an inhibitor of the tPA substrate plasmin, and receptor-associated protein (RAP), a pan-ligand blocker of the low-density lipoprotein receptor, two proteins previously reported to modulate NMDAR activity. These findings suggest that tPA can modulate changes in intracellular calcium levels in a subset of NMDARs expressed in cultured embryonic hippocampal neurons through a mechanism that involves the proteolytic activity of tPA and synaptic NMDARs.
Collapse
Affiliation(s)
- Samuel D Robinson
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand
| | - Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand
| | - David L Christie
- School of Biological Sciences and Centre for Brain Research, University of Auckland Auckland, New Zealand ; Brain Research New Zealand, Rangahau Roro Aotearoa, 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, University of Auckland Auckland, New Zealand
| |
Collapse
|
43
|
Martínez-Moreno A, Rodríguez-Durán LF, Escobar ML. Brain-derived neurotrophic factor into adult neocortex strengthens a taste aversion memory. Behav Brain Res 2015; 297:1-4. [PMID: 26433146 DOI: 10.1016/j.bbr.2015.09.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/23/2015] [Accepted: 09/25/2015] [Indexed: 11/26/2022]
Abstract
Nowadays, it is known that brain derived neurotrophic-factor (BDNF) is a protein critically involved in regulating long-term memory related mechanisms. Previous studies from our group in the insular cortex (IC), a brain structure of the temporal lobe implicated in acquisition, consolidation and retention of conditioned taste aversion (CTA), demonstrated that BDNF is essential for CTA consolidation. Recent studies show that BDNF-TrkB signaling is able to mediate the enhancement of memory. However, whether BDNF into neocortex is able to enhance aversive memories remains unexplored. In the present work, we administrated BDNF in a concentration capable of inducing in vivo neocortical LTP, into the IC immediately after CTA acquisition in two different conditions: a "strong-CTA" induced by 0.2M lithium chloride i.p. as unconditioned stimulus, and a "weak-CTA" induced by 0.1M lithium chloride i.p. Our results show that infusion of BDNF into the IC converts a weak CTA into a strong one, in a TrkB receptor-dependent manner. The present data suggest that BDNF into the adult insular cortex is sufficient to increase an aversive memory-trace.
Collapse
Affiliation(s)
- Araceli Martínez-Moreno
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, México D.F., Mexico
| | - Luis F Rodríguez-Durán
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, México D.F., Mexico
| | - Martha L Escobar
- División de Investigación y Estudios de Posgrado, Facultad de Psicología, Universidad Nacional Autónoma de México, México D.F., Mexico.
| |
Collapse
|
44
|
Tissue Plasminogen Activator Expression Is Restricted to Subsets of Excitatory Pyramidal Glutamatergic Neurons. Mol Neurobiol 2015; 53:5000-12. [PMID: 26377106 DOI: 10.1007/s12035-015-9432-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022]
Abstract
Although the extracellular serine protease tissue plasminogen activator (tPA) is involved in pathophysiological processes such as learning and memory, anxiety, epilepsy, stroke, and Alzheimer's disease, information about its regional, cellular, and subcellular distribution in vivo is lacking. In the present study, we observed, in healthy mice and rats, the presence of tPA in endothelial cells, oligodendrocytes, mastocytes, and ependymocytes, but not in pericytes, microglial cells, and astrocytes. Moreover, blockage of the axo-dendritic transport unmasked tPA expression in neurons of cortical and hippocampal areas. Interestingly, combined electrophysiological recordings, single-cell reverse transcription polymerase chain reaction (RT-PCR), and immunohistological analyses revealed that the presence of tPA is restricted to subsets of excitatory pyramidal glutamatergic neurons. We further evidenced that tPA is stored in synaptobrevin-2-positive glutamatergic synaptic vesicles. Based on all these data, we propose the existence of tPA-ergic neurons in the mature brain.
Collapse
|
45
|
Wójtowicz T, Brzdąk P, Mozrzymas JW. Diverse impact of acute and long-term extracellular proteolytic activity on plasticity of neuronal excitability. Front Cell Neurosci 2015; 9:313. [PMID: 26321914 PMCID: PMC4530619 DOI: 10.3389/fncel.2015.00313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/28/2015] [Indexed: 12/13/2022] Open
Abstract
Learning and memory require alteration in number and strength of existing synaptic connections. Extracellular proteolysis within the synapses has been shown to play a pivotal role in synaptic plasticity by determining synapse structure, function, and number. Although synaptic plasticity of excitatory synapses is generally acknowledged to play a crucial role in formation of memory traces, some components of neural plasticity are reflected by nonsynaptic changes. Since information in neural networks is ultimately conveyed with action potentials, scaling of neuronal excitability could significantly enhance or dampen the outcome of dendritic integration, boost neuronal information storage capacity and ultimately learning. However, the underlying mechanism is poorly understood. With this regard, several lines of evidence and our most recent study support a view that activity of extracellular proteases might affect information processing in neuronal networks by affecting targets beyond synapses. Here, we review the most recent studies addressing the impact of extracellular proteolysis on plasticity of neuronal excitability and discuss how enzymatic activity may alter input-output/transfer function of neurons, supporting cognitive processes. Interestingly, extracellular proteolysis may alter intrinsic neuronal excitability and excitation/inhibition balance both rapidly (time of minutes to hours) and in long-term window. Moreover, it appears that by cleavage of extracellular matrix (ECM) constituents, proteases may modulate function of ion channels or alter inhibitory drive and hence facilitate active participation of dendrites and axon initial segments (AISs) in adjusting neuronal input/output function. Altogether, a picture emerges whereby both rapid and long-term extracellular proteolysis may influence some aspects of information processing in neurons, such as initiation of action potential, spike frequency adaptation, properties of action potential and dendritic backpropagation.
Collapse
Affiliation(s)
- Tomasz Wójtowicz
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| | - Patrycja Brzdąk
- Department of Animal Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland ; Department of Animal Physiology, Institute of Experimental Biology, Wroclaw University Wroclaw, Poland
| |
Collapse
|
46
|
Tissue plasminogen activator modulates emotion in a social context. Behav Brain Res 2015; 281:24-31. [DOI: 10.1016/j.bbr.2014.11.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 11/03/2014] [Accepted: 11/08/2014] [Indexed: 11/18/2022]
|
47
|
Docagne F, Parcq J, Lijnen R, Ali C, Vivien D. Understanding the Functions of Endogenous and Exogenous Tissue-Type Plasminogen Activator During Stroke. Stroke 2015; 46:314-20. [DOI: 10.1161/strokeaha.114.006698] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Fabian Docagne
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Jérôme Parcq
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Roger Lijnen
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Carine Ali
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| | - Denis Vivien
- From the INSERM UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, Caen, France (F.D., J.P., C.A., D.V.); and Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium (R.L.)
| |
Collapse
|
48
|
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
|
49
|
Revest JM, Le Roux A, Roullot-Lacarrière V, Kaouane N, Vallée M, Kasanetz F, Rougé-Pont F, Tronche F, Desmedt A, Piazza PV. BDNF-TrkB signaling through Erk1/2 MAPK phosphorylation mediates the enhancement of fear memory induced by glucocorticoids. Mol Psychiatry 2014; 19:1001-9. [PMID: 24126929 PMCID: PMC4195976 DOI: 10.1038/mp.2013.134] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 08/27/2013] [Accepted: 08/28/2013] [Indexed: 12/25/2022]
Abstract
Activation of glucocorticoid receptors (GR) by glucocorticoid hormones (GC) enhances contextual fear memories through the activation of the Erk1/2(MAPK) signaling pathway. However, the molecular mechanism mediating this effect of GC remains unknown. Here we used complementary molecular and behavioral approaches in mice and rats and in genetically modified mice in which the GR was conditionally deleted (GR(NesCre)). We identified the tPA-BDNF-TrkB signaling pathway as the upstream molecular effectors of GR-mediated phosphorylation of Erk1/2(MAPK) responsible for the enhancement of contextual fear memory. These findings complete our knowledge of the molecular cascade through which GC enhance contextual fear memory and highlight the role of tPA-BDNF-TrkB-Erk1/2(MAPK) signaling pathways as one of the core effectors of stress-related effects of GC.
Collapse
Affiliation(s)
- J-M Revest
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France,Pathophysiology of Addiction, Neurocentre Magendie, INSERM-U862, 146 rue Léo Saignat, Bordeaux F-33077, France. E-mail:
| | - A Le Roux
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - V Roullot-Lacarrière
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - N Kaouane
- Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France,INSERM U862, Neurocentre Magendie, Pathophysiology of Declarative Memory, Bordeaux, France
| | - M Vallée
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - F Kasanetz
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - F Rougé-Pont
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| | - F Tronche
- CNRS UMR7224, UPMC Université Pierre et Marie Curie, Molecular Genetics, Neurophysiology and Behavior, Paris, France
| | - A Desmedt
- Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France,INSERM U862, Neurocentre Magendie, Pathophysiology of Declarative Memory, Bordeaux, France
| | - P V Piazza
- INSERM U862, Neurocentre Magendie, Pathophysiology of Addiction, Bordeaux, France,Pathophysiology of Neuronal Plasticity, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
50
|
Gonzalez-Gronow M, Gomez CF, de Ridder GG, Ray R, Pizzo SV. Binding of tissue-type plasminogen activator to the glucose-regulated protein 78 (GRP78) modulates plasminogen activation and promotes human neuroblastoma cell proliferation in vitro. J Biol Chem 2014; 289:25166-76. [PMID: 25059665 DOI: 10.1074/jbc.m114.589341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
The glucose-regulated protein 78 (GRP78) is a plasminogen (Pg) receptor on the cell surface. In this study, we demonstrate that GRP78 also binds the tissue-type plasminogen activator (t-PA), which results in a decrease in K(m) and an increase in the V(max) for both its amidolytic activity and activation of its substrate, Pg. This results in accelerated Pg activation when GRP78, t-PA, and Pg are bound together. The increase in t-PA activity is the result of a mechanism involving a t-PA lysine-dependent binding site in the GRP78 amino acid sequence (98)LIGRTWNDPSVQQDIKFL(115). We found that GRP78 is expressed on the surface of neuroblastoma SK-N-SH cells where it is co-localized with the voltage-dependent anion channel (VDAC), which is also a t-PA-binding protein in these cells. We demonstrate that both Pg and t-PA serve as a bridge between GRP78 and VDAC bringing them together to facilitate Pg activation. t-PA induces SK-N-SH cell proliferation via binding to GRP78 on the cell surface. Furthermore, Pg binding to the COOH-terminal region of GRP78 stimulates cell proliferation via its microplasminogen domain. This study confirms previous findings from our laboratory showing that GRP78 acts as a growth factor-like receptor and that its association with t-PA, Pg, and VDAC on the cell surface may be part of a system controlling cell growth.
Collapse
Affiliation(s)
- Mario Gonzalez-Gronow
- From the Department of Biological Sciences, Laboratory of Environmental Neurotoxicology Faculty of Medicine, Universidad Católica del Norte, Coquimbo 1781421, Chile and the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Cristian Farias Gomez
- From the Department of Biological Sciences, Laboratory of Environmental Neurotoxicology Faculty of Medicine, Universidad Católica del Norte, Coquimbo 1781421, Chile and
| | - Gustaaf G de Ridder
- the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Rupa Ray
- the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| | - Salvatore V Pizzo
- the Department of Pathology, Duke University, Medical Center, Durham, North Carolina 27710
| |
Collapse
|