1
|
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
|
2
|
Pijet B, Kostrzewska-Księzyk A, Pijet-Kucicka M, Kaczmarek L. Matrix Metalloproteinase-9 Contributes to Epilepsy Development after Ischemic Stroke in Mice. Int J Mol Sci 2024; 25:896. [PMID: 38255970 PMCID: PMC10815104 DOI: 10.3390/ijms25020896] [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: 12/07/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Epilepsy, a neurological disorder affecting over 50 million individuals globally, is characterized by an enduring predisposition and diverse consequences, both neurobiological and social. Acquired epilepsy, constituting 30% of cases, often results from brain-damaging injuries like ischemic stroke. With one third of epilepsy cases being resistant to existing drugs and without any preventive therapeutics for epileptogenesis, identifying anti-epileptogenic targets is crucial. Stroke being a leading cause of acquired epilepsy, particularly in the elderly, prompts the need for understanding post-stroke epileptogenesis. Despite the challenges in studying stroke-evoked epilepsy in rodents due to poor long-term survival rates, in this presented study the use of an animal care protocol allowed for comprehensive investigation. We highlight the role of matrix metalloproteinase-9 (MMP-9) in post-stroke epileptogenesis, emphasizing MMP-9 involvement in mouse models and its potential as a therapeutic target. Using a focal Middle Cerebral Artery occlusion model, this study demonstrates MMP-9 activation following ischemia, influencing susceptibility to seizures. MMP-9 knockout reduces epileptic features, while overexpression exacerbates them. The findings show that MMP-9 is a key player in post-stroke epileptogenesis, presenting opportunities for future therapies and expanding our understanding of acquired epilepsy.
Collapse
Affiliation(s)
- Barbara Pijet
- Laboratory of Neurobiology, Braincity, Nencki Institute of Experimental Biology, Pasteura 3, 02-093 Warsaw, Poland; (A.K.-K.)
| | | | | | | |
Collapse
|
3
|
Kuzniewska B, Rejmak K, Nowacka A, Ziółkowska M, Milek J, Magnowska M, Gruchota J, Gewartowska O, Borsuk E, Salamian A, Dziembowski A, Radwanska K, Dziembowska M. Disrupting interaction between miR-132 and Mmp9 3'UTR improves synaptic plasticity and memory in mice. Front Mol Neurosci 2022; 15:924534. [PMID: 35992198 PMCID: PMC9389266 DOI: 10.3389/fnmol.2022.924534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/18/2022] [Indexed: 11/15/2022] Open
Abstract
As microRNAs have emerged to be important regulators of molecular events occurring at the synapses, the new questions about their regulatory effect on the behavior have araised. In the present study, we show for the first time that the dysregulated specific targeting of miR132 to Mmp9 mRNA in the mouse brain results in the increased level of Mmp9 protein, which affects synaptic plasticity and has an effect on memory formation. Our data points at the importance of complex and precise regulation of the Mmp9 level by miR132 in the brain.
Collapse
Affiliation(s)
- Bozena Kuzniewska
- Laboratory of Molecular Basis of Synaptic Plasticity, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Karolina Rejmak
- Laboratory of Molecular Basis of Synaptic Plasticity, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Agata Nowacka
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Ziółkowska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Jacek Milek
- Laboratory of Molecular Basis of Synaptic Plasticity, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Marta Magnowska
- Laboratory of Molecular Basis of Synaptic Plasticity, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Jakub Gruchota
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Olga Gewartowska
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ewa Borsuk
- Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Ahmad Salamian
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Andrzej Dziembowski
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
- Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
| | - Magdalena Dziembowska
- Laboratory of Molecular Basis of Synaptic Plasticity, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| |
Collapse
|
4
|
Mercan Isik C, Uzun Cicek A, Ulger D, Bakir S. SIRT1, MMP-9 and TIMP-1 levels in children with specific learning disorder. J Psychiatr Res 2022; 152:352-359. [PMID: 35785578 DOI: 10.1016/j.jpsychires.2022.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/02/2022] [Accepted: 06/07/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Specific Learning Disorder (SLD) is a common developmental and neurobiological disorder of childhood characterized by impairment of functionality in one or more areas such as reading, writing, mathematics, listening, speaking, and reasoning. The etiology of SLD is still not fully understood. The aim of this study was to evaluate children with SLD to investigate the potential role of MMP-9, TIMP-1 and SIRT-1, which have important roles in synaptic plasticity, cognitive functions, learning and memory, and are known to be associated with various psychiatric disorders. METHODS The study was conducted with 44 outpatients aged 8-14 years who were diagnosed with SLD according to DSM-5 in the outpatient clinic and a control group of 44 age, gender and education level-matched healthy children. The groups were compared in respect of serum levels of MMP-9, TIMP-1 and SIRT-1, evaluated using the ELISA method. RESULTS Serum MMP-9 levels were significantly lower in children in the SLD group than in the control group, while TIMP-1 was higher. No difference was determined between the groups in respect of the SIRT1 levels. SLD severity was negatively correlated with MMP-9 levels and positively correlated with TIMP-1 levels. CONCLUSIONS MMP-9 appear to contribute to hippocampal-dependent memory and learning by modulating long-term synaptic plasticity. The findings of this study also reinforce the idea that deregulation of the MMP-9/TIMP-1 ratio may impact learning and play a role in SLD. These findings will help to elucidate the etiology of SLD. Furthermore, understanding molecular pathways can contribute to the discovery of certain biomarkers in SLD pathogenesis and the development of new treatment possibilities.
Collapse
Affiliation(s)
- Cansu Mercan Isik
- Department of Child and Adolescent Psychiatry, Diyarbakir Gazi Yasargil Training and Research Hospital, Diyarbakir, Turkey.
| | - Ayla Uzun Cicek
- Department of Child and Adolescent Psychiatry, Cumhuriyet University Faculty of Medicine, Sivas, Turkey.
| | - Dilara Ulger
- Department of Biochemistry, Cumhuriyet University Faculty of Medicine, Sivas, Turkey.
| | - Sevtap Bakir
- Department of Biochemistry, Cumhuriyet University Faculty of Medicine, Sivas, Turkey.
| |
Collapse
|
5
|
The association of matrix metalloproteinase 9 (MMP9) with hippocampal volume in schizophrenia: a preliminary MRI study. Neuropsychopharmacology 2022; 47:524-530. [PMID: 33833403 PMCID: PMC8674225 DOI: 10.1038/s41386-021-00997-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/13/2021] [Accepted: 03/05/2021] [Indexed: 02/02/2023]
Abstract
Matrix metalloproteinases 9 (MMP9) are enzymes involved in regulating neuroplasticity in the hippocampus. This, combined with evidence for disrupted hippocampal structure and function in schizophrenia, has prompted our current investigation into the relationship between MMP9 and hippocampal volumes in schizophrenia. 34 healthy individuals (mean age = 32.50, male = 21, female = 13) and 30 subjects with schizophrenia (mean age = 33.07, male = 19, female = 11) underwent a blood draw and T1-weighted magnetic resonance imaging. The hippocampus was automatically segmented utilizing FreeSurfer. MMP9 plasma levels were measured with ELISA. ANCOVAs were conducted to compare MMP9 plasma levels (corrected for age and sex) and hippocampal volumes between groups (corrected for age, sex, total intracranial volume). Spearman correlations were utilized to investigate the relationship between symptoms, medication, duration of illness, number of episodes, and MMP9 plasma levels in patients. Last, we explored the correlation between MMP9 levels and hippocampal volumes in patients and healthy individuals separately. Patients displayed higher MMP9 plasma levels than healthy individuals (F(1, 60) = 21.19, p < 0.0001). MMP9 levels correlated with negative symptoms in patients (R = 0.39, p = 0.035), but not with medication, duration of illness, or the number of episodes. Further, patients had smaller left (F(1,59) = 9.12, p = 0.0040) and right (F(1,59) = 6.49, p = 0.013) hippocampal volumes. Finally, left (R = -0.39, p = 0.034) and right (R = -0.37, p = 0.046) hippocampal volumes correlated negatively with MMP9 plasma levels in patients. We observe higher MMP9 plasma levels in SCZ, associated with lower hippocampal volumes, suggesting involvement of MMP9 in the pathology of SCZ. Future studies are needed to investigate how MMP9 influences the pathology of SCZ over the lifespan, whether the observed associations are specific for schizophrenia, and if a therapeutic modulation of MMP9 promotes neuroprotective effects in SCZ.
Collapse
|
6
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Menon PK, Patnaik R, Wiklund L, Sharma HS. Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury. PROGRESS IN BRAIN RESEARCH 2021; 265:139-230. [PMID: 34560921 DOI: 10.1016/bs.pbr.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Military personnel are the most susceptible to concussive head injury (CHI) caused by explosion, blast or missile or blunt head trauma. Mild to moderate CHI could induce lifetime functional and cognitive disturbances causing significant decrease in quality of life. Severe CHI leads to instant death and lifetime paralysis. Thus, further exploration of novel therapeutic agents or new features of known pharmacological agents are needed to enhance quality of life of CHI victims. Previous reports from our laboratory showed that mild CHI induced by weight drop technique causing an impact of 0.224N results in profound progressive functional deficit, memory impairment and brain pathology from 5h after trauma that continued over several weeks of injury. In this investigation we report that TiO2 nanowired delivery of oxiracetam (50mg/kg, i.p.) daily for 5 days after CHI resulted in significant improvement of functional deficit on the 8th day. This was observed using Rota Rod treadmill, memory improvement assessed by the time spent in finding hidden platform under water. The motor function improvement is seen in oxiracetam treated CHI group by placing forepaw on an inclined mesh walking and foot print analysis for stride length and distance between hind feet. TiO2-nanowired oxiracetam also induced marked improvements in the cerebral blood flow, reduction in the BBB breakdown and edema formation as well as neuroprotection of neuronal, glial and myelin damages caused by CHI at light and electron microscopy on the 7th day after 5 days TiO2 oxiracetam treatment. Adverse biochemical events such as upregulation of CSF nitrite and nitrate, IL-6, TNF-a and p-Tau are also reduced significantly in oxiracetam treated CHI group. On the other hand post treatment of 100mg/kg dose of normal oxiracetam in identical conditions after CHI is needed to show slight but significant neuroprotection together with mild recovery of memory function and functional deficits on the 8th day. These observations are the first to point out that nanowired delivery of oxiracetam has superior neuroprotective ability in CHI. These results indicate a promising clinical future of TiO2 oxiracetam in treating CHI patients for better quality of life and neurorehabilitation, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
7
|
Pulya S, Mahale A, Bobde Y, Routholla G, Patel T, Swati, Biswas S, Sharma V, Kulkarni OP, Ghosh B. PT3: A Novel Benzamide Class Histone Deacetylase 3 Inhibitor Improves Learning and Memory in Novel Object Recognition Mouse Model. ACS Chem Neurosci 2021; 12:883-892. [PMID: 33577290 DOI: 10.1021/acschemneuro.0c00721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The importance of HDAC3 in transcriptional regulation of genes associated with long-term memory is well established. Here, we report a novel HDAC3 inhibitor, PT3, with an excellent blood-brain barrier permeability and ability to enhance long-term memory in mouse model of novel object recognition (NOR). PT3 exhibited higher selectivity for HDAC3 over HDAC1, HDAC6, and HDAC8 compared to the reference compound CI994. PT3 has significant distribution into the brain tissue with Cmax at 0.5 h and t1/2 of 2.5 h. Treatment with PT3 significantly improved the discrimination index in C57/BL6 mice in the NOR model. Brain tissue analysis of mice treated with PT3 for NOR test showed significant increase in H3K9 acetylation in hippocampus. Gene expression analysis by RT-qPCR of the hippocampus tissue revealed upregulation of CREB 1, BDNF, TRKB, Nr4a2, c-fos, PKA, GAP 43, PSD 95 and MMP9 expression in mice treated with PT3. Similar to the phenotype observed in the in vivo experiment, we found upregulation of H3K9 acetylation, CREB 1, BDNF, TRKB, Nr4a2, c-fos, PKA, GAP 43 and MMP9 expression in mouse neuronal (N2A) cells treated with PT3. Thus, our preclinical studies identify PT3 as a potential HDAC3 selective inhibitor that crosses the blood-brain barrier and improves the long-term memory formation in C57/BL6 mice. We propose PT3 as a candidate with therapeutic potential to treat age-related memory loss as well as other disorders with declined memory function like Alzheimer's disease.
Collapse
Affiliation(s)
- Sravani Pulya
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Ashutosh Mahale
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Yamini Bobde
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Ganesh Routholla
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Tarun Patel
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Swati
- Department of Biological Science, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Swati Biswas
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Vivek Sharma
- Department of Biological Science, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Onkar P. Kulkarni
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| |
Collapse
|
8
|
Lenci E, Cosottini L, Trabocchi A. Novel matrix metalloproteinase inhibitors: an updated patent review (2014 - 2020). Expert Opin Ther Pat 2021; 31:509-523. [PMID: 33487088 DOI: 10.1080/13543776.2021.1881481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: Matrix MetalloProteinases (MMPs) are key enzymes in several pathophysiological processes connected to the extracellular matrix (ECM) degradation. Earlier clinical trials evaluating broad spectrum MMP inhibitors as cancer therapeutics failed to succeed, resulting in toxic side effects, such as musculoskeletal pain and inflammation, due to poor selectivity. As it is now recognized that some MMPs are essential for tumor progression and metastasis, but others play host-protective functions, selective MMP inhibitors are needed, and their interest has grown also for therapeutic applications beyond cancer, such as infectious, inflammatory and neurological diseases. Areas covered: This updated review describes patents concerning MMP inhibitors published within January 2014 and June 2020, with therapeutic applications spanning from cancer to inflammatory and neurological disorders. Expert opinion: Although the number of patents has decreased with respect to the previous decade, new applications provide selective matrix metalloproteinase inhibitors for therapeutic treatments beyond cancer. For several applications, the need of selective inhibitors resulted in the development of new non-hydroxamate compounds, paving the way towards a renewed interest towards MMPs as therapeutic targets. In particular, inhibitors able to cross the blood-brain barrier have been disclosed and proposed for the treatment of neurological conditions, infections, wound healing and cancer.
Collapse
Affiliation(s)
- Elena Lenci
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, Italy
| | - Lucrezia Cosottini
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, Italy
| | - Andrea Trabocchi
- Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
9
|
Medoro A, Bartollino S, Mignogna D, Marziliano N, Porcile C, Nizzari M, Florio T, Pagano A, Raimo G, Intrieri M, Russo C. Proteases Upregulation in Sporadic Alzheimer's Disease Brain. J Alzheimers Dis 2020; 68:931-938. [PMID: 30814362 DOI: 10.3233/jad-181284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Certain proteases are involved in Alzheimer's disease (AD) and their erroneous control may contribute to the pathology onset and progression. In this study we evaluated the cerebral expression of eight proteases, involved in both AβPP processing and extracellular matrix remodeling. Among these proteases, ADAM10, ADAMTS1, Cathepsin D, and Meprin β show a significantly higher mRNAs expression in sporadic AD subjects versus controls, while ADAMTS1, Cathepsin D, and Meprin β show an increment also at the protein level. These data indicate that transcriptional events affecting brain proteases are activated in AD patients, suggesting a link between proteolysis and AD.
Collapse
Affiliation(s)
- Alessandro Medoro
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Silvia Bartollino
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Donatella Mignogna
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Nicola Marziliano
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy.,Clinical Pathology Laboratory, ASL Taranto, Italy
| | - Carola Porcile
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Mario Nizzari
- Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Tullio Florio
- Department of Internal Medicine and Centre of Excellence for Biomedical Research, University of Genova, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Aldo Pagano
- Department of Experimental Medicine, University of Genova, Genova, Italy.,IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gennaro Raimo
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Mariano Intrieri
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Claudio Russo
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
10
|
Kalpachidou T, Makrygiannis AK, Pavlakis E, Stylianopoulou F, Chalepakis G, Stamatakis A. Behavioural effects of extracellular matrix protein Fras1 depletion in the mouse. Eur J Neurosci 2020; 53:3905-3919. [DOI: 10.1111/ejn.14759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/21/2020] [Accepted: 04/17/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Theodora Kalpachidou
- Biology‐Biochemistry Lab Faculty of Nursing School of Health Sciences National and Kapodistrian University of Athens Athens Greece
| | | | | | - Fotini Stylianopoulou
- Biology‐Biochemistry Lab Faculty of Nursing School of Health Sciences National and Kapodistrian University of Athens Athens Greece
| | | | - Antonios Stamatakis
- Biology‐Biochemistry Lab Faculty of Nursing School of Health Sciences National and Kapodistrian University of Athens Athens Greece
| |
Collapse
|
11
|
Venlafaxine Stimulates an MMP-9-Dependent Increase in Excitatory/Inhibitory Balance in a Stress Model of Depression. J Neurosci 2020; 40:4418-4431. [PMID: 32269106 DOI: 10.1523/jneurosci.2387-19.2020] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 11/21/2022] Open
Abstract
Emerging evidence suggests that there is a reduction in overall cortical excitatory to inhibitory balance in major depressive disorder (MDD), which afflicts ∼14%-20% of individuals. Reduced pyramidal cell arborization occurs with stress and MDD, and may diminish excitatory neurotransmission. Enhanced deposition of perineuronal net (PNN) components also occurs with stress. Since parvalbumin-expressing interneurons are the predominant cell population that is enveloped by PNNs, which enhance their ability to release GABA, excess PNN deposition likely increases pyramidal cell inhibition. In the present study, we investigate the potential for matrix metalloprotease-9 (MMP-9), an endopeptidase secreted in response to neuronal activity, to contribute to the antidepressant efficacy of the serotonin/norepinephrine reuptake inhibitor venlafaxine in male mice. Chronic venlafaxine increases MMP-9 levels in murine cortex, and increases both pyramidal cell arborization and PSD-95 expression in the cortex of WT but not MMP-9-null mice. We have previously shown that venlafaxine reduces PNN deposition and increases the power of ex vivo γ oscillations in conventionally housed mice. γ power is increased with pyramidal cell disinhibition and with remission from MDD. Herein we observe that PNN expression is increased in a corticosterone-induced stress model of disease and reduced by venlafaxine. Compared with mice that receive concurrent venlafaxine, corticosterone-treated mice also display reduced ex vivo γ power and impaired working memory. Autopsy-derived PFC samples show elevated MMP-9 levels in antidepressant-treated MDD patients compared with controls. These preclinical and postmortem findings highlight a link between extracellular matrix regulation and MDD.SIGNIFICANCE STATEMENT Reduced excitatory neurotransmission occurs with major depressive disorder, and may be normalized by antidepressant treatment. Underlying molecular mechanisms are, however, not well understood. Herein we investigate a potential role for an extracellular protease, released from neurons and known to play a role in learning and memory, in antidepressant-associated increases in excitatory transmission. Our data suggest that this protease, matrix metalloprotease-9, increases branching of excitatory neurons and concomitantly attenuates the perineuronal net to potentially reduce inhibitory input to these neurons. Matrix metalloprotease-9 may thus enhance overall excitatory/inhibitory balance and neuronal population dynamics, which are important to mood and memory.
Collapse
|
12
|
Kaminari A, Tsilibary EC, Tzinia A. A New Perspective in Utilizing MMP-9 as a Therapeutic Target for Alzheimer's Disease and Type 2 Diabetes Mellitus. J Alzheimers Dis 2019; 64:1-16. [PMID: 29865065 DOI: 10.3233/jad-180035] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Matrix metalloprotease 9 (MMP-9) is a 92 kDa type IV collagenase and a member of the family of endopeptidases. MMP-9 is involved in the degradation of extracellular matrix components, tissue remodeling, cellular receptor stripping, and processing of various signaling molecules. In the CNS, the effects of MMP-9 are quite complex, since it exerts beneficial effects including neurogenesis, angiogenesis, myelogenesis, axonal growth, and inhibition of apoptosis, or destructive effects including apoptosis, blood-brain barrier disorder, and demyelination. Likewise, in the periphery, physiological events, as the involvement of MMP-9 in angiogenesis, for instance in wound healing, can be turned into pathological, such as in tumor metastasis, depending on the state of the organism. Alzheimer's disease is a neurodegenerative disorder, characterized by amyloid accumulation and deposition in the brain. Amyloidogenesis, however, also occurs in diseases of the periphery, such as type II diabetes mellitus, where an analogous type of amyloid, is deposited in the pancreas. Interestingly, both diseases exhibit similar pathology and disease progression, with insulin resistance being a major common denominator. Hence, combinatorial strategies searching new or existing molecules to apply for therapeutic use for both diseases are gaining momentum. MMP-9 is extensively studied due to its association with a variety of physiological and pathological processes. Consequently, meticulous design could render MMP-9 into a potential therapeutic target for Alzheimer's disease and type 2 diabetes mellitus; two seemingly unrelated diseases.
Collapse
Affiliation(s)
- Archontia Kaminari
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Effie C Tsilibary
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Athina Tzinia
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| |
Collapse
|
13
|
Alaiyed S, Conant K. A Role for Matrix Metalloproteases in Antidepressant Efficacy. Front Mol Neurosci 2019; 12:117. [PMID: 31133801 PMCID: PMC6517485 DOI: 10.3389/fnmol.2019.00117] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/24/2019] [Indexed: 01/10/2023] Open
Abstract
Major depressive disorder is a debilitating condition that affects approximately 15% of the United States population. Though the neurophysiological mechanisms that underlie this disorder are not completely understood, both human and rodent studies suggest that excitatory/inhibitory (E/I) balance is reduced with the depressive phenotype. In contrast, antidepressant efficacy in responsive individuals correlates with increased excitatory neurotransmission in select brain regions, suggesting that the restoration of E/I balance may improve mood. Enhanced excitatory transmission can occur through mechanisms including increased dendritic arborization and synapse formation in pyramidal neurons. Reduced activity of inhibitory neurons may also contribute to antidepressant efficacy. Consistent with this possibility, the fast-acting antidepressant ketamine may act by selective inhibition of glutamatergic input to GABA releasing parvalbumin (PV)-expressing interneurons. Recent work has also shown that a negative allosteric modulator of the GABA-A receptor α subunit can improve depression-related behavior. PV-expressing interneurons are thought to represent critical pacemakers for synchronous network events. These neurons also represent the predominant GABAergic neuronal population that is enveloped by the perineuronal net (PNN), a lattice-like structure that is thought to stabilize glutamatergic input to this cell type. Disruption of the PNN reduces PV excitability and increases pyramidal cell excitability. Various antidepressant medications increase the expression of matrix metalloproteinases (MMPs), enzymes that can increase pyramidal cell dendritic arborization and spine formation. MMPs can also cleave PNN proteins to reduce PV neuron-mediated inhibition. The present review will focus on mechanisms that may underlie antidepressant efficacy, with a focus on monoamines as facilitators of increased matrix metalloprotease (MMP) expression and activation. Discussion will include MMP-dependent effects on pyramidal cell structure and function, as well as MMP-dependent effects on PV expressing interneurons. We conclude with discussion of antidepressant use for those at risk for Alzheimer’s disease, and we also highlight areas for further study.
Collapse
Affiliation(s)
- Seham Alaiyed
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC, United States
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
14
|
Nucleic acid-induced potentiation of matrix metalloproteinase-9 enzymatic activity. Biochem J 2018; 475:1597-1610. [PMID: 29654109 PMCID: PMC5941315 DOI: 10.1042/bcj20180035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 12/31/2022]
Abstract
Matrix metalloproteinases (MMPs) play varied roles in normal biology and diseases where, depending on the context, both inhibition and enhancement of the enzymatic activity may be beneficial. However, there are very few reports of positive modulators of MMP activity. We report that polynucleotides, including single-stranded DNA, RNA, and even double-stranded DNA, bind to and enhance the enzymatic activity of MMP9. This enhancement of MMP9 catalytic activity is not shared by biologically active polycationic molecules suggesting nonspecific charge screening as an unlikely mechanism. Deletion construct and MMP1, 2, and 3 studies suggest that the type-II fibronectin repeat domains of the enzyme appear to play a role in mediating the nucleotide potentiation of MMP9 activity. Single-stranded DNA enhances nerve growth factor-induced MMP9-dependent neurite extension in pheochromocytoma 12 cells providing evidence for potential biological significance of the nucleotide-mediated allosteric enhancement of the catalytic activity.
Collapse
|
15
|
Pijet B, Stefaniuk M, Kostrzewska-Ksiezyk A, Tsilibary PE, Tzinia A, Kaczmarek L. Elevation of MMP-9 Levels Promotes Epileptogenesis After Traumatic Brain Injury. Mol Neurobiol 2018; 55:9294-9306. [PMID: 29667129 PMCID: PMC6208832 DOI: 10.1007/s12035-018-1061-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/03/2018] [Indexed: 12/24/2022]
Abstract
Posttraumatic epilepsy (PTE) is a recurrent seizure disorder that often develops secondary to traumatic brain injury (TBI) that is caused by an external mechanical force. Recent evidence shows that the brain extracellular matrix plays a major role in the remodeling of neuronal connections after injury. One of the proteases that is presumably responsible for this process is matrix metalloproteinase-9 (MMP-9). The levels of MMP-9 are elevated in rodent brain tissue and human blood samples after TBI. However, no studies have described the influence of MMP-9 on the development of PTE. The present study used controlled cortical impact (CCI) as a mouse model of TBI. We examined the detailed kinetics of MMP-9 levels for 1 month after TBI and observed two peaks after injury (30 min and 6 h after injury). We tested the hypothesis that high levels of MMP-9 predispose individuals to the development of PTE, and MMP-9 inhibition would protect against PTE. We used transgenic animals with either MMP-9 knockout or MMP-9 overexpression. MMP-9 overexpression increased the number of mice that exhibited TBI-induced spontaneous seizures, and MMP-9 knockout decreased the appearance of seizures. We also evaluated changes in responsiveness to a single dose of the chemoconvulsant pentylenetetrazol. MMP-9-overexpressing mice exhibited a significantly shorter latency between pentylenetetrazol administration and the first epileptiform spike. MMP-9 knockout mice exhibited the opposite response profile. Finally, we found that the occurrence of PTE was correlated with the size of the lesion after injury. Overall, our data emphasize the contribution of MMP-9 to TBI-induced structural and physiological alterations in brain circuitry that may lead to the development of PTE.
Collapse
Affiliation(s)
- Barbara Pijet
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Pasteura 3, 02-093, Warsaw, Poland.
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Pasteura 3, 02-093, Warsaw, Poland
| | - Agnieszka Kostrzewska-Ksiezyk
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Pasteura 3, 02-093, Warsaw, Poland
| | - Photini-Effie Tsilibary
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, 55405, USA.,Brain Sciences Center, Minneapolis, MN, 55417, USA
| | - Athina Tzinia
- Laboratory of Cell and Matrix Pathobiology, Institute of Bioscience and Applications, NCSR Demokritos, 153 10 Aghia Paraskevi Attikis, Athens, Greece
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
16
|
Biella G, Fusco F, Nardo E, Bernocchi O, Colombo A, Lichtenthaler SF, Forloni G, Albani D. Sirtuin 2 Inhibition Improves Cognitive Performance and Acts on Amyloid-β Protein Precursor Processing in Two Alzheimer's Disease Mouse Models. J Alzheimers Dis 2018; 53:1193-207. [PMID: 27372638 DOI: 10.3233/jad-151135] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The neuropathological hallmarks of Alzheimer's disease (AD) are extracellular plaques built up by the accumulation of the amyloid-β protein precursor (AβPP)-derived peptide β (Aβ), and intracellular tangles of hyperphosphorylated tau protein. Sirtuin 2 (SIRT2) is a member of the sirtuin family, featuring conserved enzymes with deacetylase activity and involved in several cell molecular pathways. We investigated the importance of SIRT2 inhibition in AD. We inhibited SIRT2 by small molecules (AGK-2, AK-7) and examined AβPP metabolism in H4-SW neuroglioma cells overexpressing AβPP and two AD transgenic mouse models (3xTg-AD and APP23). The in vitro studies suggested that the inhibition of SIRT2 reduced Aβ production; in vivo data showed an improvement of cognitive performance in the novel object recognition test, and an effect on AβPP proteolytic processing leading to a reduction of soluble β-AβPP and an increase of soluble α-AβPP protein. In 3xTg-AD mice, we noticed that total tau protein level rose. Overall, our pre-clinical data support a role for SIRT2 inhibition in the improvement of cognitive performance and the modulation of molecular mechanisms relevant for AD, thus deserving attention as possible therapeutic strategy.
Collapse
Affiliation(s)
- Gloria Biella
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Milan, Italy
| | - Federica Fusco
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Milan, Italy
| | - Emanuele Nardo
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Milan, Italy
| | - Ottavia Bernocchi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Milan, Italy
| | - Alessio Colombo
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München, Munich, Germany.,Munich Center for Systems Neurology (SyNergy), Munich, Germany
| | - Gianluigi Forloni
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Milan, Italy
| | - Diego Albani
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Milan, Italy
| |
Collapse
|
17
|
Yin J, Liu C, Shen Y, Zhang H, Cao J. Efficacy of ursolic acid against Echinococcus granulosus in vitro and in a murine infection model. Parasit Vectors 2018; 11:58. [PMID: 29368624 PMCID: PMC5784668 DOI: 10.1186/s13071-018-2628-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 01/08/2018] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cystic echinococcosis is a global public health problem; however, the drugs (albendazole and mebendazole) currently recommended by WHO for its treatment, have limited efficacy. Therefore, novel drugs are required to provide more choices for the treatment of this disease. METHODS The anthelmintic effects of ursolic acid (UA) were tested on Echinococcus granulosus protoscoleces, germinal cells and metacestodes in vitro. The in vivo efficacy of UA was investigated in mice following secondary infection with E. granulosus. Furthermore, the corresponding ultrastructural damage induced by UA was evaluated by electron microscopy. RESULTS In vitro, 45.95 ± 5.30% of protoscoleces were killed by UA at 40 μg/ml, while the growth of more than 90% of germinal cells was inhibited by UA at 10 to 40 μg/ml. The same effect was observed in metacestodes 7 days after treatment with UA at 10, 20 and 40 μg/ml, and more than 50% of metacestodes showed loss of integrity at the end of the experiment. In vivo, metacestode weight was significantly reduced following oral administration of UA at 200 and 100 mg/kg (39.5 and 38.3%, respectively). Additionally, ultrastructural damage, such as alternations in germinal cell morphology and formation of vacuoles and lipid granules were observed in parasites treated with UA in vitro, while detachment of the germinal layer from the laminated layer was also seen in metacestodes in vivo. CONCLUSIONS UA was demonstrated to exert parasiticidal activity against E. granulosus in vitro and in vivo, thus implicating UA as a potential anti-echinococcosis agent.
Collapse
Affiliation(s)
- Jianhai Yin
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China
- Key Laboratory of Parasite and Vector Biology, MOH, Shanghai, 200025 China
- National Center for International Research on Tropical Diseases, Shanghai, 200025 China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025 China
| | - Congshan Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China
- Key Laboratory of Parasite and Vector Biology, MOH, Shanghai, 200025 China
- National Center for International Research on Tropical Diseases, Shanghai, 200025 China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025 China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China
- Key Laboratory of Parasite and Vector Biology, MOH, Shanghai, 200025 China
- National Center for International Research on Tropical Diseases, Shanghai, 200025 China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025 China
| | - Haobing Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China
- Key Laboratory of Parasite and Vector Biology, MOH, Shanghai, 200025 China
- National Center for International Research on Tropical Diseases, Shanghai, 200025 China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025 China
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China
- Key Laboratory of Parasite and Vector Biology, MOH, Shanghai, 200025 China
- National Center for International Research on Tropical Diseases, Shanghai, 200025 China
- WHO Collaborating Centre for Tropical Diseases, Shanghai, 200025 China
| |
Collapse
|
18
|
Brzdak P, Nowak D, Wiera G, Mozrzymas JW. Multifaceted Roles of Metzincins in CNS Physiology and Pathology: From Synaptic Plasticity and Cognition to Neurodegenerative Disorders. Front Cell Neurosci 2017; 11:178. [PMID: 28713245 PMCID: PMC5491558 DOI: 10.3389/fncel.2017.00178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) and membrane proteolysis play a key role in structural and functional synaptic plasticity associated with development and learning. A growing body of evidence underscores the multifaceted role of members of the metzincin superfamily, including metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs), A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTSs) and astacins in physiological and pathological processes in the central nervous system (CNS). The expression and activity of metzincins are strictly controlled at different levels (e.g., through the regulation of translation, limited activation in the extracellular space, the binding of endogenous inhibitors and interactions with other proteins). Thus, unsurprising is that the dysregulation of proteolytic activity, especially the greater expression and activation of metzincins, is associated with neurodegenerative disorders that are considered synaptopathies, especially Alzheimer's disease (AD). We review current knowledge of the functions of metzincins in the development of AD, mainly the proteolytic processing of amyloid precursor protein, the degradation of amyloid β (Aβ) peptide and several pathways for Aβ clearance across brain barriers (i.e., blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB)) that contain specific receptors that mediate the uptake of Aβ peptide. Controlling the proteolytic activity of metzincins in Aβ-induced pathological changes in AD patients' brains may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Patrycja Brzdak
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Daria Nowak
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Grzegorz Wiera
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| |
Collapse
|
19
|
De Luca C, Papa M. Matrix Metalloproteinases, Neural Extracellular Matrix, and Central Nervous System Pathology. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:167-202. [PMID: 28662822 DOI: 10.1016/bs.pmbts.2017.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The functionality and stability of the central nervous system (CNS) pabulum, called neural extracellular matrix (nECM), is paramount for the maintenance of a healthy network. The loosening or the damage of the scaffold disrupts synaptic transmission with the consequent imbalance of the neurotransmitters, reactive cells invasion, astrocytosis, new matrix deposition, digestion of the previous structure and ultimately, maladaptive plasticity with the loss of neuronal viability. nECM is constantly affected by CNS disorders, particularly in chronic modifying such as neurodegenerative disease, or in acute/subacute with chronic sequelae, like cerebrovascular and inflammatory pathology. Matrix metalloproteinases (MMPs) are the main interfering agent of nECM, guiding the balance of degradation and new deposition of proteins such as proteoglycans and glycoproteins, or glycosaminoglycans, such as hyaluronic acid. Activation of these enzymes is modulated by their physiologic inhibitors, the tissue inhibitors of MMPs or via other proteases inhibitors, as well as genetic or epigenetic up- or downregulation through molecular interaction or receptor activation. The appropriate understanding of the pathways underlying nECM modifications in CNS pathology is probably one of the pivotal future directions to identify the healthy brain network and subsequently design new therapies to interfere with the progression of the CNS disease and eventually find appropriate therapies.
Collapse
Affiliation(s)
- Ciro De Luca
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Papa
- Laboratory of Neuronal Networks, University of Campania "Luigi Vanvitelli", Naples, Italy; SYSBIO, Centre for Systems Biology, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
20
|
Overexpression of matrix metalloproteinase-9 (MMP-9) rescues insulin-mediated impairment in the 5XFAD model of Alzheimer's disease. Sci Rep 2017; 7:683. [PMID: 28386117 PMCID: PMC5429641 DOI: 10.1038/s41598-017-00794-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/13/2017] [Indexed: 12/30/2022] Open
Abstract
A hallmark of Alzheimer’s disease (AD) is the accumulation of oligomeric amyloid-β (Aβ) peptide, which may be primarily responsible for neuronal dysfunction. Insulin signaling provides a defense mechanism against oligomer-induced neuronal loss. We previously described the neuroprotective role of matrix metalloproteinase 9 (MMP-9) in decreasing the formation of Aβ oligomers. In the present study, we examined the role of MMP-9 on the insulin survival pathway in primary hippocampal cultures and hippocampal cell extracts from 3 month-old wild type, AD (5XFAD), MMP-9-overexpressing (TgMMP-9), and double transgenic mice (5XFAD/TgMMP-9). The data demonstrate that the insulin pathway was compromised in samples from 5XFAD mice, when compared to the wild type and TgMMP-9. This was due to enhanced phosphorylation of IRS1 at Serine 636 (pIRS1-Ser636), which renders IRS1 inactive and prevents insulin-mediated signaling. In 5XFAD/TgMMP-9 samples, the insulin survival pathway was rescued through enhanced activation by phosphorylation of IRS1 at Tyrosine 465 (pIRS1-Tyr465), downstream increased phosphorylation of Akt and GSK-3β, and decreased phosphorylation of JNK kinase. Oligomeric Aβ levels decreased and BDNF levels increased in 5XFAD/TgMMP-9 mice, compared to 5XFAD mice. Our findings indicate that overexpression of MMP-9 rescued insulin survival signaling in vitro and in early stages in the 5XFAD model of AD.
Collapse
|
21
|
Tan WH, Bird LM. Angelman syndrome: Current and emerging therapies in 2016. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2016; 172:384-401. [PMID: 27860204 DOI: 10.1002/ajmg.c.31536] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally-inherited UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS) at the unmethylated paternal locus. We reviewed all published information on the clinical trials that have been completed as well as the publicly available information on ongoing trials of therapies in AS. To date, all clinical trials that strove to improve neurodevelopment in AS have been unsuccessful. Attempts at hypermethylating the maternal locus through dietary compounds were ineffective. The results of an 8-week open-label trial using minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive, while a subsequent randomized placebo-controlled trial suggested that treatment with minocycline for 8 weeks did not result in any neurodevelopmental gains. A 1-year randomized placebo-controlled trial using levodopa to alter the phosphorylation of calcium/calmodulin-dependent kinase II did not lead to any improvement in neurodevelopment. Topoisomerase inhibitors and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Artificial transcription factors are being developed to "super activate" UBE3A or inhibit UBE3A-AS. Other strategies targeting specific pathways are briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances, which are two of the more common complications of AS. © 2016 Wiley Periodicals, Inc.
Collapse
|
22
|
Lebida K, Mozrzymas JW. Spike Timing-Dependent Plasticity in the Mouse Barrel Cortex Is Strongly Modulated by Sensory Learning and Depends on Activity of Matrix Metalloproteinase 9. Mol Neurobiol 2016; 54:6723-6736. [PMID: 27744572 PMCID: PMC5622912 DOI: 10.1007/s12035-016-0174-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Abstract
Experience and learning in adult primary somatosensory cortex are known to affect neuronal circuits by modifying both excitatory and inhibitory transmission. Synaptic plasticity phenomena provide a key substrate for cognitive processes, but precise description of the cellular and molecular correlates of learning is hampered by multiplicity of these mechanisms in various projections and in different types of neurons. Herein, we investigated the impact of associative learning on neuronal plasticity in distinct types of postsynaptic neurons by checking the impact of classical conditioning (pairing whisker stroking with tail shock) on the spike timing-dependent plasticity (t-LTP and t-LTD) in the layer IV to II/III vertical pathway of the mouse barrel cortex. Learning in this paradigm practically prevented t-LTP measured in pyramidal neurons but had no effect on t-LTD. Since classical conditioning is known to affect inhibition in the barrel cortex, we examined its effect on tonic GABAergic currents and found a strong downregulation of these currents in the layer II/III interneurons but not in pyramidal cells. Matrix metalloproteinases emerged as crucial players in synaptic plasticity and learning. We report that the blockade of MMP-9 (but not MMP-3) abolished t-LTP having no effect on t-LTD. Moreover, associative learning resulted in an upregulation of gelatinolytic activity within the "trained" barrel. We conclude that LTP induced by spike timing-dependent plasticity (STDP) paradigm is strongly correlated with associative learning and critically depends on the activity of MMP-9.
Collapse
Affiliation(s)
- Katarzyna Lebida
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, Chalubinskiego 3a, 50-368, Wroclaw, Poland.
| | - Jerzy W Mozrzymas
- Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University, Chalubinskiego 3a, 50-368, Wroclaw, Poland.,Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| |
Collapse
|
23
|
Fisher JR, Wallace CE, Tripoli DL, Sheline YI, Cirrito JR. Redundant Gs-coupled serotonin receptors regulate amyloid-β metabolism in vivo. Mol Neurodegener 2016; 11:45. [PMID: 27315796 PMCID: PMC4912779 DOI: 10.1186/s13024-016-0112-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 06/14/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The aggregation of amyloid-β (Aβ) into insoluble plaques is a hallmark pathology of Alzheimer's disease (AD). Previous work has shown increasing serotonin levels with selective serotonin re-uptake inhibitor (SSRI) compounds reduces Aβ in the brain interstitial fluid (ISF) in a mouse model of AD and in the cerebrospinal fluid of humans. We investigated which serotonin receptor (5-HTR) subtypes and downstream effectors were responsible for this reduction. RESULTS Agonists of 5-HT4R, 5-HT6R, and 5-HT7R significantly reduced ISF Aβ, but agonists of other receptor subtypes did not. Additionally, inhibition of Protein Kinase A (PKA) blocked the effects of citalopram, an SSRI, on ISF Aβ levels. Serotonin signaling does not appear to change gene expression to reduce Aβ levels in acute timeframes, but likely acts within the cytoplasm to increase α-secretase enzymatic activity. Broad pharmacological inhibition of putative α-secretases increased ISF Aβ and blocked the effects of citalopram. CONCLUSIONS In total, these studies map the major signaling components linking serotonin receptors to suppression of brain ISF Aβ. These results suggest the reduction in ISF Aβ is mediated by a select group of 5-HTRs and open future avenues for targeted therapy of AD.
Collapse
Affiliation(s)
- Jonathan R Fisher
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Clare E Wallace
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Danielle L Tripoli
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Yvette I Sheline
- Departments of Psychiatry, Radiology, and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - John R Cirrito
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA. .,Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA. .,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA. .,Present Address: Washington University, Neurology, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO, 63110, USA.
| |
Collapse
|
24
|
Vafadari B, Salamian A, Kaczmarek L. MMP-9 in translation: from molecule to brain physiology, pathology, and therapy. J Neurochem 2016; 139 Suppl 2:91-114. [PMID: 26525923 DOI: 10.1111/jnc.13415] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) is a member of the metzincin family of mostly extracellularly operating proteases. Despite the fact that all of these enzymes might be target promiscuous, with largely overlapping catalogs of potential substrates, MMP-9 has recently emerged as a major and apparently unique player in brain physiology and pathology. The specificity of MMP-9 may arise from its very local and time-restricted actions, even when released in the brain from cells of various types, including neurons, glia, and leukocytes. In fact, the quantity of MMP-9 is very low in the naive brain, but it is markedly activated at the levels of enzymatic activity, protein abundance, and gene expression following various physiological stimuli and pathological insults. Neuronal MMP-9 participates in synaptic plasticity by controlling the shape of dendritic spines and function of excitatory synapses, thus playing a pivotal role in learning, memory, and cortical plasticity. When improperly unleashed, MMP-9 contributes to a large variety of brain disorders, including epilepsy, schizophrenia, autism spectrum disorder, brain injury, stroke, neurodegeneration, pain, brain tumors, etc. The foremost mechanism of action of MMP-9 in brain disorders appears to be its involvement in immune/inflammation responses that are related to the enzyme's ability to process and activate various cytokines and chemokines, as well as its contribution to blood-brain barrier disruption, facilitating the extravasation of leukocytes into brain parenchyma. However, another emerging possibility (i.e., the control of MMP-9 over synaptic plasticity) should not be neglected. The translational potential of MMP-9 has already been recognized in both the diagnosis and treatment domains. The most striking translational aspect may be the discovery of MMP-9 up-regulation in a mouse model of Fragile X syndrome, quickly followed by human studies and promising clinical trials that have sought to inhibit MMP-9. With regard to diagnosis, suggestions have been made to use MMP-9 alone or combined with tissue inhibitor of matrix metalloproteinase-1 or brain-derived neurotrophic factor as disease biomarkers. MMP-9, through cleavage of specific target proteins, plays a major role in synaptic plasticity and neuroinflammation, and by those virtues contributes to brain physiology and a host of neurological and psychiatric disorders. This article is part of the 60th Anniversary special issue.
Collapse
|
25
|
Pharmacological therapies for Angelman syndrome. Wien Med Wochenschr 2016; 167:205-218. [PMID: 26758979 DOI: 10.1007/s10354-015-0408-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/15/2015] [Indexed: 12/16/2022]
Abstract
Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by a loss of the maternally inherited UBE3A; the paternal UBE3A is silenced in neurons by a mechanism involving an antisense transcript (UBE3A-AS). We reviewed the published information on clinical trials that have been completed as well as the publicly available information on ongoing trials of therapies for AS. Attempts at hypermethylating the maternal locus through dietary compounds were ineffective. The results of a clinical trial using minocycline as a matrix metalloproteinase-9 inhibitor were inconclusive; another clinical trial is underway. Findings from a clinical trial using L-dopa to alter phosphorylation of calcium/calmodulin-dependent kinase II are awaited. Topoisomerase inhibitors and antisense oligonucleotides are being developed to directly inhibit UBE3A-AS. Other strategies targeting specific pathways are briefly discussed. We also reviewed the medications that are currently used to treat seizures and sleep disturbances, which are two of the more debilitating manifestations of AS.
Collapse
|
26
|
Baranger K, Marchalant Y, Bonnet AE, Crouzin N, Carrete A, Paumier JM, Py NA, Bernard A, Bauer C, Charrat E, Moschke K, Seiki M, Vignes M, Lichtenthaler SF, Checler F, Khrestchatisky M, Rivera S. MT5-MMP is a new pro-amyloidogenic proteinase that promotes amyloid pathology and cognitive decline in a transgenic mouse model of Alzheimer's disease. Cell Mol Life Sci 2016; 73:217-36. [PMID: 26202697 PMCID: PMC4700096 DOI: 10.1007/s00018-015-1992-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 01/22/2023]
Abstract
Membrane-type 5-matrix metalloproteinase (MT5-MMP) is a proteinase mainly expressed in the nervous system with emerging roles in brain pathophysiology. The implication of MT5-MMP in Alzheimer's disease (AD), notably its interplay with the amyloidogenic process, remains elusive. Accordingly, we crossed the genetically engineered 5xFAD mouse model of AD with MT5-MMP-deficient mice and examined the impact of MT5-MMP deficiency in bigenic 5xFAD/MT5-MMP(-/-) mice. At early stages (4 months) of the pathology, the levels of amyloid beta peptide (Aβ) and its amyloid precursor protein (APP) C-terminal fragment C99 were largely reduced in the cortex and hippocampus of 5xFAD/MT5-MMP(-/-), compared to 5xFAD mice. Reduced amyloidosis in bigenic mice was concomitant with decreased glial reactivity and interleukin-1β (IL-1β) levels, and the preservation of long-term potentiation (LTP) and spatial learning, without changes in the activity of α-, β- and γ-secretases. The positive impact of MT5-MMP deficiency was still noticeable at 16 months of age, as illustrated by reduced amyloid burden and gliosis, and a better preservation of the cortical neuronal network and synaptophysin levels in bigenic mice. MT5-MMP expressed in HEKswe cells colocalized and co-immunoprecipitated with APP and significantly increased the levels of Aβ and C99. MT5-MMP also promoted the release of a soluble APP fragment of 95 kDa (sAPP95) in HEKswe cells. sAPP95 levels were significantly reduced in brain homogenates of 5xFAD/MT5-MMP(-/-) mice, supporting altogether the idea that MT5-MMP influences APP processing. MT5-MMP emerges as a new pro-amyloidogenic regulator of APP metabolism, whose deficiency alleviates amyloid pathology, neuroinflammation and cognitive decline.
Collapse
Affiliation(s)
- Kévin Baranger
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Yannick Marchalant
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
- Psychology Department, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Amandine E Bonnet
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Nadine Crouzin
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Alex Carrete
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | | | - Nathalie A Py
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Anne Bernard
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Charlotte Bauer
- Labex DistAlz, IPMC UMR 7275 CNRS-UNS, 06560, Valbonne, France
| | - Eliane Charrat
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France
| | - Katrin Moschke
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Munich, Germany
- Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM), 81675, Munich, Germany
| | - Mothoharu Seiki
- Division of Cancer Cell Research, Institute of Medical Science, University of Tokyo, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Michel Vignes
- UMR5247 IBMM CNRS University of Montpellier 1 and University of Montpellier 2, 34095, Montepellier, France
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE) and Neuroproteomics, Munich, Germany
- Klinikum rechts der Isar, and Institute for Advanced Study, Technische Universität München (TUM), 81675, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
| | | | | | - Santiago Rivera
- Aix-Marseille Université, CNRS, NICN UMR 7259, 13344, Marseille, France.
| |
Collapse
|
27
|
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: 1.9] [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]
|
28
|
Abstract
Recent findings implicate alterations in glutamate signaling, leading to aberrant synaptic plasticity, in schizophrenia. Matrix metalloproteinase-9 (MMP-9) has been shown to regulate glutamate receptors, be regulated by glutamate at excitatory synapses, and modulate physiological and morphological synaptic plasticity. By means of functional gene polymorphism, gene responsiveness to antipsychotics and blood plasma levels MMP-9 has recently been implicated in schizophrenia. This commentary critically reviews these findings based on the hypothesis that MMP-9 contributes to pathological synaptic plasticity in schizophrenia.
Collapse
Affiliation(s)
- Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Warsaw, Poland
| | - Leszek Kaczmarek
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Warsaw, Poland
| |
Collapse
|
29
|
Rochais C, Lecoutey C, Gaven F, Giannoni P, Hamidouche K, Hedou D, Dubost E, Genest D, Yahiaoui S, Freret T, Bouet V, Dauphin F, Sopkova de Oliveira Santos J, Ballandonne C, Corvaisier S, Malzert-Fréon A, Legay R, Boulouard M, Claeysen S, Dallemagne P. Novel multitarget-directed ligands (MTDLs) with acetylcholinesterase (AChE) inhibitory and serotonergic subtype 4 receptor (5-HT4R) agonist activities as potential agents against Alzheimer's disease: the design of donecopride. J Med Chem 2015; 58:3172-87. [PMID: 25793650 DOI: 10.1021/acs.jmedchem.5b00115] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In this work, we describe the synthesis and in vitro evaluation of a novel series of multitarget-directed ligands (MTDL) displaying both nanomolar dual-binding site (DBS) acetylcholinesterase inhibitory effects and partial 5-HT4R agonist activity, among which donecopride was selected for further in vivo evaluations in mice. The latter displayed procognitive and antiamnesic effects and enhanced sAPPα release, accounting for a potential symptomatic and disease-modifying therapeutic benefit in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Christophe Rochais
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Cédric Lecoutey
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Florence Gaven
- ‡CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.,§Inserm, U1191, F-34000 Montpellier, France.,∥Université de Montpellier, UMR-5203, F-34000 Montpellier, France
| | - Patrizia Giannoni
- ‡CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.,§Inserm, U1191, F-34000 Montpellier, France.,∥Université de Montpellier, UMR-5203, F-34000 Montpellier, France
| | - Katia Hamidouche
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Damien Hedou
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Emmanuelle Dubost
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - David Genest
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Samir Yahiaoui
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Thomas Freret
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Valentine Bouet
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - François Dauphin
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | | | - Céline Ballandonne
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Sophie Corvaisier
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France.,⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Aurélie Malzert-Fréon
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Remi Legay
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Michel Boulouard
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Sylvie Claeysen
- ‡CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.,§Inserm, U1191, F-34000 Montpellier, France.,∥Université de Montpellier, UMR-5203, F-34000 Montpellier, France
| | - Patrick Dallemagne
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| |
Collapse
|
30
|
Filali M, Lalonde R. Motor activity in young APPswe + PS1/A246E bigenic mice as a predicting variable for memory decline. J Neurosci Res 2015; 93:948-53. [PMID: 25594937 DOI: 10.1002/jnr.23552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/27/2014] [Accepted: 12/02/2014] [Indexed: 12/19/2022]
Abstract
Reports of individuality in rodent species have been a subject of debate in pharmacology and other fields. In the current study, APPswe + PS1/A246E bigenic mice with Alzheimer's-like pathogenesis and wild-type controls were subdivided at 3 months of age into low, intermediate, and high responders in open-field activity. The mice were then evaluated longitudinally at 3 and 9 months for object recognition. Irrespective of genotype, mice with a high level of motor activity had better scores in object recognition. However, a significant correlation was established between open-field activity measured at 3 months of age and recognition memory measured at 9 months of age in the bigenic group only. These results indicate that motor activity in young mice with amyloid neuropathology may serve as a predicting variable for cognitive dysfunction in more mature mice.
Collapse
Affiliation(s)
- Mohammed Filali
- Functional Analysis of Animal Behavior Platform, CHU de Québec Research Center, and Department of Molecular Medicine, Laval University, Québec City, Québec, Canada
| | | |
Collapse
|
31
|
Stargardt A, Swaab DF, Bossers K. The storm before the quiet: neuronal hyperactivity and Aβ in the presymptomatic stages of Alzheimer's disease. Neurobiol Aging 2015; 36:1-11. [DOI: 10.1016/j.neurobiolaging.2014.08.014] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/29/2014] [Accepted: 08/12/2014] [Indexed: 12/27/2022]
|
32
|
Gkogkas CG, Khoutorsky A, Cao R, Jafarnejad SM, Prager-Khoutorsky M, Giannakas N, Kaminari A, Fragkouli A, Nader K, Price TJ, Konicek BW, Graff JR, Tzinia AK, Lacaille JC, Sonenberg N. Pharmacogenetic inhibition of eIF4E-dependent Mmp9 mRNA translation reverses fragile X syndrome-like phenotypes. Cell Rep 2014; 9:1742-1755. [PMID: 25466251 PMCID: PMC4294557 DOI: 10.1016/j.celrep.2014.10.064] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 09/30/2014] [Accepted: 10/24/2014] [Indexed: 01/22/2023] Open
Abstract
Fragile X syndrome (FXS) is the leading genetic cause of autism. Mutations in Fmr1 (fragile X mental retardation 1 gene) engender exaggerated translation resulting in dendritic spine dysmorphogenesis, synaptic plasticity alterations, and behavioral deficits in mice, which are reminiscent of FXS phenotypes. Using postmortem brains from FXS patients and Fmr1 knockout mice (Fmr1(-/y)), we show that phosphorylation of the mRNA 5' cap binding protein, eukaryotic initiation factor 4E (eIF4E), is elevated concomitant with increased expression of matrix metalloproteinase 9 (MMP-9) protein. Genetic or pharmacological reduction of eIF4E phosphorylation rescued core behavioral deficits, synaptic plasticity alterations, and dendritic spine morphology defects via reducing exaggerated translation of Mmp9 mRNA in Fmr1(-/y) mice, whereas MMP-9 overexpression produced several FXS-like phenotypes. These results uncover a mechanism of regulation of synaptic function by translational control of Mmp-9 in FXS, which opens the possibility of new treatment avenues for the diverse neurological and psychiatric aspects of FXS.
Collapse
Affiliation(s)
- Christos G Gkogkas
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada.
| | - Arkady Khoutorsky
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada
| | - Ruifeng Cao
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada
| | - Seyed Mehdi Jafarnejad
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada
| | - Masha Prager-Khoutorsky
- Center for Research in Neuroscience, McGill University, Montréal General Hospital, Montréal, QC H3G 1A4, Canada
| | - Nikolaos Giannakas
- Institute of Biosciences and Applications, National Center for Scientific Research Demokritos, Agia Paraskevi, 15310 Athens, Greece
| | - Archontia Kaminari
- Institute of Biosciences and Applications, National Center for Scientific Research Demokritos, Agia Paraskevi, 15310 Athens, Greece
| | - Apostolia Fragkouli
- Institute of Biosciences and Applications, National Center for Scientific Research Demokritos, Agia Paraskevi, 15310 Athens, Greece
| | - Karim Nader
- Department of Psychology, McGill University, Montréal, QC H3A 1B1, Canada
| | - Theodore J Price
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Bruce W Konicek
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Jeremy R Graff
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Athina K Tzinia
- Institute of Biosciences and Applications, National Center for Scientific Research Demokritos, Agia Paraskevi, 15310 Athens, Greece
| | - Jean-Claude Lacaille
- GRSNC and Department of Neurosciences, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada.
| |
Collapse
|
33
|
Transcriptional evidence for the role of chronic venlafaxine treatment in neurotrophic signaling and neuroplasticity including also Glutamatergic [corrected] - and insulin-mediated neuronal processes. PLoS One 2014; 9:e113662. [PMID: 25423262 PMCID: PMC4244101 DOI: 10.1371/journal.pone.0113662] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 10/27/2014] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVES Venlafaxine (VLX), a serotonine-noradrenaline reuptake inhibitor, is one of the most commonly used antidepressant drugs in clinical practice for the treatment of major depressive disorder (MDD). Despite being more potent than its predecessors, similarly to them, the therapeutical effect of VLX is visible only 3-4 weeks after the beginning of treatment. Furthermore, recent papers show that antidepressants, including also VLX, enhance the motor recovery after stroke even in non depressed persons. In the present, transcriptomic-based study we looked for changes in gene expressions after a long-term VLX administration. METHODS Osmotic minipumps were implanted subcutaneously into Dark Agouti rats providing a continuous (40 mg/kg/day) VLX delivery for three weeks. Frontal regions of the cerebral cortex were isolated and analyzed using Illumina bead arrays to detect genes showing significant chances in expression. Gene set enrichment analysis was performed to identify specific regulatory networks significantly affected by long term VLX treatment. RESULTS Chronic VLX administration may have an effect on neurotransmitter release via the regulation of genes involved in vesicular exocytosis and receptor endocytosis (such as Kif proteins, Myo5a, Sv2b, Syn2 or Synj2). Simultaneously, VLX activated the expression of genes involved in neurotrophic signaling (Ntrk2, Ntrk3), glutamatergic transmission (Gria3, Grin2b and Grin2a), neuroplasticity (Camk2g/b, Cd47), synaptogenesis (Epha5a, Gad2) and cognitive processes (Clstn2). Interestingly, VLX increased the expression of genes involved in mitochondrial antioxidant activity (Bcl2 and Prdx1). Additionally, VLX administration also modulated genes related to insulin signaling pathway (Negr1, Ppp3r1, Slc2a4 and Enpp1), a mechanism that has recently been linked to neuroprotection, learning and memory. CONCLUSIONS Our results strongly suggest that chronic VLX treatment improves functional reorganization and brain plasticity by influencing gene expression in regulatory networks of motor cortical areas. These results are consonant with the synaptic (network) hypothesis of depression and antidepressant-induced motor recovery after stroke.
Collapse
|
34
|
Mukherjee A, Swarnakar S. Implication of matrix metalloproteinases in regulating neuronal disorder. Mol Biol Rep 2014; 42:1-11. [DOI: 10.1007/s11033-014-3752-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
35
|
Fragkouli A, Tsilibary EC, Tzinia AK. Neuroprotective role of MMP-9 overexpression in the brain of Alzheimer's 5xFAD mice. Neurobiol Dis 2014; 70:179-89. [DOI: 10.1016/j.nbd.2014.06.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/05/2014] [Accepted: 06/28/2014] [Indexed: 12/20/2022] Open
|
36
|
Lee YS. Genes and signaling pathways involved in memory enhancement in mutant mice. Mol Brain 2014; 7:43. [PMID: 24894914 PMCID: PMC4050447 DOI: 10.1186/1756-6606-7-43] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 05/27/2014] [Indexed: 11/10/2022] Open
Abstract
Mutant mice have been used successfully as a tool for investigating the mechanisms of memory at multiple levels, from genes to behavior. In most cases, manipulating a gene expressed in the brain impairs cognitive functions such as memory and their underlying cellular mechanisms, including synaptic plasticity. However, a remarkable number of mutations have been shown to enhance memory in mice. Understanding how to improve a system provides valuable insights into how the system works under normal conditions, because this involves understanding what the crucial components are. Therefore, more can be learned about the basic mechanisms of memory by studying mutant mice with enhanced memory. This review will summarize the genes and signaling pathways that are altered in the mutants with enhanced memory, as well as their roles in synaptic plasticity. Finally, I will discuss how knowledge of memory-enhancing mechanisms could be used to develop treatments for cognitive disorders associated with impaired plasticity.
Collapse
Affiliation(s)
- Yong-Seok Lee
- Department of Life Science, College of Natural Science, Chung-Ang University, Seoul 156-756, Republic of Korea.
| |
Collapse
|
37
|
Juan WS, Huang SY, Chang CC, Hung YC, Lin YW, Chen TY, Lee AH, Lee AC, Wu TS, Lee EJ. Melatonin improves neuroplasticity by upregulating the growth-associated protein-43 (GAP-43) and NMDAR postsynaptic density-95 (PSD-95) proteins in cultured neurons exposed to glutamate excitotoxicity and in rats subjected to transient focal cerebral ischemia even during a long-term recovery period. J Pineal Res 2014; 56:213-23. [PMID: 24350898 DOI: 10.1111/jpi.12114] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 12/13/2013] [Indexed: 01/22/2023]
Abstract
Recent evidence shows that the NMDAR postsynaptic density-95 (PSD-95), growth-associated protein-43 (GAP-43), and matrix metalloproteinase-9 (MMP-9) protein enhance neuroplasticity at the subacute stage of stroke. Here, we evaluated whether melatonin would modulate the PSD-95, GAP-43, and MMP-9 proteins in cultured neurons exposed to glutamate excitotoxicity and in rats subjected to experimental stroke. Adult male Sprague-Dawley rats were treated with melatonin (5 mg/kg) or vehicle at reperfusion onset after transient occlusion of the right middle cerebral artery (tMCAO) for 90 min. Animals were euthanized for Western immunoblot analyses for the PSD-95 and GAP-43 proteins and gelatin zymography for the MMP-9 activity at 7 days postinsult. Another set of animals was sacrificed for histologic and Golgi-Cox-impregnated sections at 28 days postinsult. In cultured neurons exposed to glutamate excitotoxicity, melatonin significantly upregulated the GAP-43 and PSD-95 expressions and improved dendritic aborizations (P<0.05, respectively). Relative to controls, melatonin-treated stroke animals caused a significant improvement in GAP-43 and PSD-95 expressions as well as the MMP-9 activity in the ischemic brain (P<0.05). Consequently, melatonin also significantly promoted the dendritic spine density and reduced infarction in the ischemic brain, and improved neurobehaviors as well at 28 days postinsult (P<0.05, respectively). Together, melatonin upregulates GAP-43, PSD-95, and MMP-9 proteins, which likely accounts for its actions to improve neuroplasticity in cultured neurons exposed to glutamate excitotoxicity and to enhance long-term neuroprotection, neuroplasticity, and brain remodeling in stroke rats.
Collapse
Affiliation(s)
- Wei-Sheng Juan
- Neurophysiology Laboratory, Institute of Biomedical Engineering & Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hernandez-Rapp J, Martin-Lannerée S, Hirsch TZ, Launay JM, Mouillet-Richard S. Hijacking PrP(c)-dependent signal transduction: when prions impair Aβ clearance. Front Aging Neurosci 2014; 6:25. [PMID: 24592237 PMCID: PMC3938157 DOI: 10.3389/fnagi.2014.00025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Indexed: 01/29/2023] Open
Abstract
The cellular prion protein PrPc is the normal counterpart of the scrapie prion protein PrP Sc, the main component of the infectious agent of transmissible spongiform encephalopathies. The recent discovery that PrP c can serve as a receptor for the amyloid beta (Aβ) peptide and relay its neurotoxicity is sparking renewed interest on this protein and its involvement in signal transduction processes. Disease-associated PrP Sc shares with Aβ the ability to hijack PrP c-dependent signaling cascades, and thereby instigate pathogenic events. Among these is an impairment of Aβ clearance, uncovered in prion-infected neuronal cells. These findings add another facet to the intricate interplay between PrP c and Aβ. Here, we summarize the connection between PrP-mediated signaling and Aβ clearance and discuss its pathological implications.
Collapse
Affiliation(s)
- Julia Hernandez-Rapp
- INSERM UMR-S1124 Paris, France ; Sorbonne Paris Cité, UMR-S1124, Université Paris Descartes Paris, France ; Université Paris Sud 11, ED419 Biosigne Orsay, France
| | - Séverine Martin-Lannerée
- INSERM UMR-S1124 Paris, France ; Sorbonne Paris Cité, UMR-S1124, Université Paris Descartes Paris, France
| | - Théo Z Hirsch
- INSERM UMR-S1124 Paris, France ; Sorbonne Paris Cité, UMR-S1124, Université Paris Descartes Paris, France
| | - Jean-Marie Launay
- AP-HP Service de Biochimie, Fondation FondaMental, INSERM U942 H ôpital Lariboisière Paris, France ; Pharma Research Department, F. Hoffmann-La-Roche Ltd. Basel, Switzerland
| | - Sophie Mouillet-Richard
- INSERM UMR-S1124 Paris, France ; Sorbonne Paris Cité, UMR-S1124, Université Paris Descartes Paris, France
| |
Collapse
|
39
|
Morawski M, Filippov M, Tzinia A, Tsilibary E, Vargova L. ECM in brain aging and dementia. PROGRESS IN BRAIN RESEARCH 2014; 214:207-27. [PMID: 25410360 DOI: 10.1016/b978-0-444-63486-3.00010-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An essential component of the brain extracellular space is the extracellular matrix contributing to the spatial assembly of cells by binding cell-surface adhesion molecules, supporting cell migration, differentiation, and tissue development. The most interesting and complex functions of the central nervous system are the abilities to encode new information (learning) and to store this information (memory). The creation of perineuronal nets, consisting mostly of chondroitin sulfate proteoglycans, stabilizes the synapses and memory trails and forms protective shields against neurodegenerative processes but terminates plasticity and the potential for recovery of the tissue. Age-related changes in the extracellular matrix composition and the extracellular space volume and permissivity are major determinants of the onset and development of the most common neurodegenerative disorder, Alzheimer's disease. In this regard, heparan sulfate proteoglycans, involved in amyloid clearance from the brain, play an important role in Alzheimer's disease and other types of neurodegeneration. Additional key players in the modification of the extracellular matrix are matrix metalloproteinases. Recent studies show that the extracellular matrix and matrix metalloproteinases are important regulators of plasticity, learning, and memory and might be involved in different neurological disorders like epilepsy, schizophrenia, addiction, and dementia. The identification of molecules and mechanisms that modulate these processes is crucial for the understanding of brain function and dysfunction and for the design of new therapeutic approaches targeting the molecular mechanism underlying these neurological disorders.
Collapse
Affiliation(s)
- Markus Morawski
- University of Leipzig, EU-ESF Transnational Junior Research Group "MESCAMP", Paul Flechsig Institute for Brain Research, Leipzig, Germany.
| | - Mikhail Filippov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Athina Tzinia
- NCSR "Demokritos", Institute of Biosciences and Applications, Athens, Greece
| | - Effie Tsilibary
- NCSR "Demokritos", Institute of Biosciences and Applications, Athens, Greece
| | - Lydia Vargova
- Charles University, 2nd Faculty of Medicine, Department of Neuroscience, Prague, Czech Republic; Institute of Experimental Medicine AS CR, v.v.i., Department of Neuroscience, Prague, Czech Republic
| |
Collapse
|
40
|
Lee H, Lee EJ, Song YS, Kim E. Long-term depression-inducing stimuli promote cleavage of the synaptic adhesion molecule NGL-3 through NMDA receptors, matrix metalloproteinases and presenilin/γ-secretase. Philos Trans R Soc Lond B Biol Sci 2013; 369:20130158. [PMID: 24298159 PMCID: PMC3843889 DOI: 10.1098/rstb.2013.0158] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Long-term depression (LTD) reduces the functional strength of excitatory synapses through mechanisms that include the removal of AMPA glutamate receptors from the postsynaptic membrane. LTD induction is also known to result in structural changes at excitatory synapses, including the shrinkage of dendritic spines. Synaptic adhesion molecules are thought to contribute to the development, function and plasticity of neuronal synapses largely through their trans-synaptic adhesions. However, little is known about how synaptic adhesion molecules are altered during LTD. We report here that NGL-3 (netrin-G ligand-3), a postsynaptic adhesion molecule that trans-synaptically interacts with the LAR family of receptor tyrosine phosphatases and intracellularly with the postsynaptic scaffolding protein PSD-95, undergoes a proteolytic cleavage process. NGL-3 cleavage is induced by NMDA treatment in cultured neurons and low-frequency stimulation in brain slices and requires the activities of NMDA glutamate receptors, matrix metalloproteinases (MMPs) and presenilin/γ-secretase. These results suggest that NGL-3 is a novel substrate of MMPs and γ-secretase and that NGL-3 cleavage may regulate synaptic adhesion during LTD.
Collapse
Affiliation(s)
- Hyejin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), , Daejeon 305-701, Korea
| | | | | | | |
Collapse
|
41
|
Soleman S, Filippov MA, Dityatev A, Fawcett JW. Targeting the neural extracellular matrix in neurological disorders. Neuroscience 2013; 253:194-213. [PMID: 24012743 DOI: 10.1016/j.neuroscience.2013.08.050] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/06/2013] [Accepted: 08/26/2013] [Indexed: 01/15/2023]
Abstract
The extracellular matrix (ECM) is known to regulate important processes in neuronal cell development, activity and growth. It is associated with the structural stabilization of neuronal processes and synaptic contacts during the maturation of the central nervous system. The remodeling of the ECM during both development and after central nervous system injury has been shown to affect neuronal guidance, synaptic plasticity and their regenerative responses. Particular interest has focused on the inhibitory role of chondroitin sulfate proteoglycans (CSPGs) and their formation into dense lattice-like structures, termed perineuronal nets (PNNs), which enwrap sub-populations of neurons and restrict plasticity. Recent studies in mammalian systems have implicated CSPGs and PNNs in regulating and restricting structural plasticity. The enzymatic degradation of CSPGs or destabilization of PNNs has been shown to enhance neuronal activity and plasticity after central nervous system injury. This review focuses on the role of the ECM, CSPGs and PNNs; and how developmental and pharmacological manipulation of these structures have enhanced neuronal plasticity and aided functional recovery in regeneration, stroke, and amblyopia. In addition to CSPGs, this review also points to the functions and potential therapeutic value of these and several other key ECM molecules in epileptogenesis and dementia.
Collapse
Affiliation(s)
- S Soleman
- Cambridge Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
42
|
Lacoste B, Tong XK, Lahjouji K, Couture R, Hamel E. Cognitive and cerebrovascular improvements following kinin B1 receptor blockade in Alzheimer's disease mice. J Neuroinflammation 2013; 10:57. [PMID: 23642031 PMCID: PMC3710240 DOI: 10.1186/1742-2094-10-57] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 04/20/2013] [Indexed: 12/11/2022] Open
Abstract
Background Recent evidence suggests that the inducible kinin B1 receptor (B1R) contributes to pathogenic neuroinflammation induced by amyloid-beta (Aβ) peptide. The present study aims at identifying the cellular distribution and potentially detrimental role of B1R on cognitive and cerebrovascular functions in a mouse model of Alzheimer’s disease (AD). Methods Transgenic mice overexpressing a mutated form of the human amyloid precursor protein (APPSwe,Ind, line J20) were treated with a selective and brain penetrant B1R antagonist (SSR240612, 10 mg/kg/day for 5 or 10 weeks) or vehicle. The impact of B1R blockade was measured on i) spatial learning and memory performance in the Morris water maze, ii) cerebral blood flow (CBF) responses to sensory stimulation using laser Doppler flowmetry, and iii) reactivity of isolated cerebral arteries using online videomicroscopy. Aβ burden was quantified by ELISA and immunostaining, while other AD landmarks were measured by western blot and immunohistochemistry. Results B1R protein levels were increased in APP mouse hippocampus and, prominently, in reactive astrocytes surrounding Aβ plaques. In APP mice, B1R antagonism with SSR240612 improved spatial learning, memory and normalized protein levels of the memory-related early gene Egr-1 in the dentate gyrus of the hippocampus. B1R antagonism restored sensory-evoked CBF responses, endothelium-dependent dilations, and normalized cerebrovascular protein levels of endothelial nitric oxide synthase and B2R. In addition, SSR240612 reduced (approximately 50%) microglial, but not astroglial, activation, brain levels of soluble Aβ1-42, diffuse and dense-core Aβ plaques, and it increased protein levels of the Aβ brain efflux transporter lipoprotein receptor-related protein-1 in cerebral microvessels. Conclusion These findings show a selective upregulation of astroglial B1R in the APP mouse brain, and the capacity of the B1R antagonist to abrogate amyloidosis, cerebrovascular and memory deficits. Collectively, these findings provide convincing evidence for a role of B1R in AD pathogenesis.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | | | | | | | | |
Collapse
|
43
|
Verslegers M, Lemmens K, Van Hove I, Moons L. Matrix metalloproteinase-2 and -9 as promising benefactors in development, plasticity and repair of the nervous system. Prog Neurobiol 2013; 105:60-78. [PMID: 23567503 DOI: 10.1016/j.pneurobio.2013.03.004] [Citation(s) in RCA: 310] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/01/2013] [Accepted: 03/28/2013] [Indexed: 11/16/2022]
Abstract
It has been 50 years since Gross and Lapiere discovered collagenolytic activity during tadpole tail metamorphosis, which was later on revealed as MMP-1, the founding member of the matrix metalloproteinases (MMPs). Currently, MMPs constitute a large group of endoproteases that are not only able to cleave all protein components of the extracellular matrix, but also to activate or inactivate many other signaling molecules, such as receptors, adhesion molecules and growth factors. Elevated MMP levels are associated with an increasing number of injuries and disorders, such as cancer, inflammation and auto-immune diseases. Yet, MMP upregulation has also been implicated in many physiological functions such as embryonic development, wound healing and angiogenesis and therefore, these proteinases are considered to be crucial mediators in many biological processes. Over the past decennia, MMP research has gained considerable attention in several pathologies, most prominently in the field of cancer metastasis, and more recent investigations also focus on the nervous system, with a striking emphasis on the gelatinases, MMP-2 and MMP-9. Unfortunately, the contribution of these gelatinases to neuropathological disorders, like multiple sclerosis and Alzheimer's disease, has overshadowed their potential as modulators of fundamental nervous system functions. Within this review, we wish to highlight the currently known or suggested actions of MMP-2 and MMP-9 in the developing and adult nervous system and their potential to improve repair or regeneration after nervous system injury.
Collapse
Affiliation(s)
- Mieke Verslegers
- Laboratory of Neural Circuit Development and Regeneration, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
44
|
Wiera G, Wozniak G, Bajor M, Kaczmarek L, Mozrzymas JW. Maintenance of long-term potentiation in hippocampal mossy fiber-CA3 pathway requires fine-tuned MMP-9 proteolytic activity. Hippocampus 2013; 23:529-43. [DOI: 10.1002/hipo.22112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2013] [Indexed: 01/08/2023]
|
45
|
Filali M, Lalonde R. The effects of subchronic d-serine on left–right discrimination learning, social interaction, and exploratory activity in APPswe/PS1 mice. Eur J Pharmacol 2013; 701:152-8. [DOI: 10.1016/j.ejphar.2012.12.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/12/2012] [Accepted: 12/19/2012] [Indexed: 10/27/2022]
|
46
|
Kostomoiri M, Fragkouli A, Sagnou M, Skaltsounis LA, Pelecanou M, Tsilibary EC, Tzinia AK. Oleuropein, an anti-oxidant polyphenol constituent of olive promotes α-secretase cleavage of the amyloid precursor protein (AβPP). Cell Mol Neurobiol 2013; 33:147-54. [PMID: 23053546 DOI: 10.1007/s10571-012-9880-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/22/2012] [Indexed: 01/09/2023]
Abstract
Over the past decade, intense focus has been dedicated on investigating processes involved in the proteolysis of amyloid precursor protein (AβPP) and β-amyloid (Aβ) peptide metabolism, as possible targets for Alzheimer's disease (AD) therapy. To this goal, considerable research has been targeted on potential therapeutic use of compounds promoting non-amyloidogenic processing of AβPP. One of these compounds, oleuropein, a polyphenol constituent of extra virgin olive oil exhibiting a wide range of pharmacological properties, was shown to interact non-covalently with Aβ, an interaction that might be related to a potential protective role of oleuropein against Aβ aggregation. In the present study, it was demonstrated that oleuropein treatment of HEK293 cells stably transfected with the isoform 695 of human AβPP (APP695) leads to markedly elevated levels of sAPPα and to significant reduction of Aβ oligomers. These effects were associated with increased activity of matrix metalloproteinase 9 (MMP-9), whereas no significant alterations in the expression of secretases TACE, ADAM-10 or BACE-1 were observed. Similar results were obtained using the human neuroblastoma cell line SK-N-SH. The experimental data reveal an anti-amyloidogenic effect of oleuropein and suggest a possible protective role for oleuropein against AD, extending the spectrum of beneficial properties of this naturally occurring polyphenol.
Collapse
Affiliation(s)
- Myrta Kostomoiri
- Institute of Biology, National Centre for Scientific Research Demokritos, 15310, Agia Paraskevi, Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
47
|
Impairment of novel object recognition in adulthood after neonatal exposure to diazinon. Arch Toxicol 2012; 87:753-62. [PMID: 23212306 DOI: 10.1007/s00204-012-0989-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 11/21/2012] [Indexed: 01/04/2023]
Abstract
Diazinon is an organophosphate pesticide that is still heavily used in agriculture, home gardening, and indoor pest control in Japan. The present study investigated the effect of neonatal exposure to diazinon on hippocampus-dependent novel object recognition test performance and the expression of the N-methyl-D-aspartate (NMDA) receptor and its signal transduction pathway-related genes in the hippocampi of young adult and adult mice. Male offspring of C3H/HeN mice were subcutaneously treated with 0, 0.5, or 5 mg/kg of diazinon for 4 consecutive days beginning on postnatal day (PND) 8. Beginning on PND 46 or PND 81, a novel object recognition test was performed on 4 consecutive days. The hippocampi were collected on PND 50 or PND 85 after the completion of the novel object recognition test, and the expression levels of neurotrophins and the NMDA receptor and its signal transduction pathway-related genes were examined using real-time RT-PCR. Diazinon-injected mice exhibited a poor ability to discriminate between novel and familiar objects during both the PND 49 and the PND 84 tests. The NMDA receptor subunits NR1 and NR2B and the related protein kinase calcium/calmodulin-dependent protein kinase (CaMK)-IV and the transcription factor cyclic AMP responsive element binding protein (CREB)-1 mRNA levels were reduced in the PND 50 mice. However, no significant changes in the expressions of the NMDA subunits and their signal transduction molecules were observed in the hippocampi of the PND 85 mice. The expression level of nerve growth factor mRNA was significantly reduced in the PND 50 or 85 mice. These results indicate that neonatal diazinon exposure impaired the hippocampus-dependent novel object recognition ability, accompanied by a modulation in the expressions of the NMDA receptor and neurotrophin in young adult and adult mice.
Collapse
|
48
|
Huntley GW. Synaptic circuit remodelling by matrix metalloproteinases in health and disease. Nat Rev Neurosci 2012; 13:743-57. [PMID: 23047773 PMCID: PMC4900464 DOI: 10.1038/nrn3320] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Matrix metalloproteinases (MMPs) are extracellularly acting enzymes that have long been known to have deleterious roles in brain injury and disease. In particular, widespread and protracted MMP activity can contribute to neuronal loss and synaptic dysfunction. However, recent studies show that rapid and focal MMP-mediated proteolysis proactively drives synaptic structural and functional remodelling that is crucial for ongoing cognitive processes. Deficits in synaptic remodelling are associated with psychiatric and neurological disorders, and aberrant MMP expression or function may contribute to the molecular mechanisms underlying these deficits. This Review explores the paradigm shift in our understanding of the contribution of MMPs to normal and abnormal synaptic plasticity and function.
Collapse
Affiliation(s)
- George W Huntley
- Fishberg Department of Neuroscience, Friedman Brain Institute and the Graduate School of Biological Sciences, The Mount Sinai School of Medicine, New York, New York 10029, USA.
| |
Collapse
|
49
|
Niedringhaus M, Chen X, Dzakpasu R, Conant K. MMPs and soluble ICAM-5 increase neuronal excitability within in vitro networks of hippocampal neurons. PLoS One 2012; 7:e42631. [PMID: 22912716 PMCID: PMC3418258 DOI: 10.1371/journal.pone.0042631] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 07/09/2012] [Indexed: 12/16/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases that are released from neurons in an activity dependent manner. Published studies suggest their activity is important to varied forms of learning and memory. At least one MMP can stimulate an increase in the size of dendritic spines, structures which represent the post synaptic component for a large number of glutamatergic synapses. This change may be associated with increased synaptic glutamate receptor incorporation, and an increased amplitude and/or frequency of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) mini excitatory post-synaptic currents (EPSCs). An associated increase in the probability of action potential occurrence would be expected. While the mechanism(s) by which MMPs may influence synaptic structure and function are not completely understood, MMP dependent shedding of specific cell adhesion molecules (CAMs) could play an important role. CAMs are ideally positioned to be cleaved by synaptically released MMPs, and shed N terminal domains could potentially interact with previously unengaged integrins to stimulate dendritic actin polymerization with spine expansion. In the present study, we have used multielectrode arrays (MEAs) to investigate MMP and soluble CAM dependent changes in neuronal activity recorded from hippocampal cultures. We have focused on intercellular adhesion molecule-5 (ICAM-5) in particular, as this CAM is expressed on glutamatergic dendrites and shed in an MMP dependent manner. We show that chemical long-term potentiation (cLTP) evoked changes in recorded activity, and the dynamics of action potential bursts in particular, are altered by MMP inhibition. A blocking antibody to β1 integrins has a similar effect. We also show that the ectodomain of ICAM-5 can stimulate β1 integrin dependent increases in spike counts and burst number. These results support a growing body of literature suggesting that MMPs have important effects on neuronal excitability. They also support the possibility that MMP dependent shedding of specific synaptic CAMs can contribute to these effects.
Collapse
Affiliation(s)
- Mark Niedringhaus
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, District of Columbia, United States of America
| | - Rhonda Dzakpasu
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- Department of Physics, Georgetown University, Washington, District of Columbia, United States of America
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- * E-mail: (KC); (RD)
| | - Katherine Conant
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, United States of America
- * E-mail: (KC); (RD)
| |
Collapse
|
50
|
Win-Shwe TT, Fujimaki H, Fujitani Y, Hirano S. Novel object recognition ability in female mice following exposure to nanoparticle-rich diesel exhaust. Toxicol Appl Pharmacol 2012; 262:355-62. [PMID: 22659509 DOI: 10.1016/j.taap.2012.05.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/17/2012] [Accepted: 05/23/2012] [Indexed: 01/21/2023]
Abstract
Recently, our laboratory reported that exposure to nanoparticle-rich diesel exhaust (NRDE) for 3 months impaired hippocampus-dependent spatial learning ability and up-regulated the expressions of memory function-related genes in the hippocampus of female mice. However, whether NRDE affects the hippocampus-dependent non-spatial learning ability and the mechanism of NRDE-induced neurotoxicity was unknown. Female BALB/c mice were exposed to clean air, middle-dose NRDE (M-NRDE, 47 μg/m(3)), high-dose NRDE (H-NRDE, 129 μg/m(3)), or filtered H-NRDE (F-DE) for 3 months. We then investigated the effect of NRDE exposure on non-spatial learning ability and the expression of genes related to glutamate neurotransmission using a novel object recognition test and a real-time RT-PCR analysis, respectively. We also examined microglia marker Iba1 immunoreactivity in the hippocampus using immunohistochemical analyses. Mice exposed to H-NRDE or F-DE could not discriminate between familiar and novel objects. The control and M-NRDE-exposed groups showed a significantly increased discrimination index, compared to the H-NRDE-exposed group. Although no significant changes in the expression levels of the NMDA receptor subunits were observed, the expression of glutamate transporter EAAT4 was decreased and that of glutamic acid decarboxylase GAD65 was increased in the hippocampus of H-NRDE-exposed mice, compared with the expression levels in control mice. We also found that microglia activation was prominent in the hippocampal area of the H-NRDE-exposed mice, compared with the other groups. These results indicated that exposure to NRDE for 3 months impaired the novel object recognition ability. The present study suggests that genes related to glutamate metabolism may be involved in the NRDE-induced neurotoxicity observed in the present mouse model.
Collapse
Affiliation(s)
- Tin-Tin Win-Shwe
- Center for Environmental Health Sciences, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan.
| | | | | | | |
Collapse
|