1
|
Catto F, Kirschenbaum D, Economides AE, Reuss AM, Trevisan C, Caredio D, Dadgar-Kiani E, Mirzet D, Frick L, Weber-Stadlbauer U, Litvinov S, Koumoutsakos P, Lee JH, Aguzzi A. Quantitative 3D histochemistry reveals region-specific amyloid-β reduction by the antidiabetic drug netoglitazone. PLoS One 2025; 20:e0309489. [PMID: 40327707 PMCID: PMC12054868 DOI: 10.1371/journal.pone.0309489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/29/2025] [Indexed: 05/08/2025] Open
Abstract
A hallmark of Alzheimer's disease (AD) is the extracellular aggregation of toxic amyloid-beta (Aβ) peptides in form of plaques. Here, we identify netoglitazone, an antidiabetic compound previously tested in humans, as an Aβ aggregation antagonist. Netoglitazone improved cognition and reduced microglia activity in a mouse model of AD. Using quantitative whole-brain three-dimensional histology (Q3D), we precisely identified brain regions where netoglitazone reduced the number and size of Aβ plaques. We demonstrate the utility of Q3D in preclinical drug evaluation for AD by providing a high-resolution brain-wide view of drug efficacy. Applying Q3D has the potential to improve pre-clinical drug evaluation by providing information that can help identify mechanisms leading to brain region-specific drug efficacy.
Collapse
Affiliation(s)
- Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- IMAI MedTech GmbH, Zurich, Switzerland
| | - Daniel Kirschenbaum
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athena E. Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Davide Caredio
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ehsan Dadgar-Kiani
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Delic Mirzet
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukas Frick
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, United States of America
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, United States of America
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, California, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Department of Electrical Engineering, Stanford University, Stanford, California, United States of America
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
3
|
Liu Z, Zhang J, Li S, Wang H, Ren B, Li J, Bao Z, Liu J, Guo M, Yang G, Chen L. Circadian control of ConA-induced acute liver injury and inflammatory response via Bmal1 regulation of Junb. JHEP Rep 2023; 5:100856. [PMID: 37791375 PMCID: PMC10542646 DOI: 10.1016/j.jhepr.2023.100856] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 10/05/2023] Open
Abstract
Background & Aims Circadian rhythms play significant roles in immune responses, and many inflammatory processes in liver diseases are associated with malfunctioning molecular clocks. However, the significance of the circadian clock in autoimmune hepatitis (AIH), which is characterised by immune-mediated hepatocyte destruction and extensive inflammatory cytokine production, remains unclear. Methods We tested the difference in susceptibility to the immune-mediated liver injury induced by concanavalin A (ConA) at various time points throughout a day in mice and analysed the effects of global, hepatocyte, or myeloid cell deletion of the core clock gene, Bmal1 (basic helix-loop-helix ARNT-like 1), on liver injury and inflammatory responses. Multiple molecular biology techniques and mice with macrophage-specific knockdown of Junb, a Bmal1 target gene, were used to investigate the involvement of Junb in the circadian control of ConA-induced hepatitis. Results The susceptibility to ConA-induced liver injury is highly dependent on the timing of ConA injection. The treatment at Zeitgeber time 0 (lights on) triggers the highest mortality as well as the severest liver injury and inflammatory responses. Further study revealed that this timing effect was driven by macrophage, but not hepatocyte, Bmal1. Mechanistically, Bmal1 controls the diurnal variation of ConA-induced hepatitis by directly regulating the circadian transcription of Junb and promoting M1 macrophage activation. Inhibition of Junb in macrophages blunts the administration time-dependent effect of ConA and attenuates liver injury. Moreover, we demonstrated that Junb promotes macrophage inflammation by regulating AKT and extracellular signal-regulated kinase (ERK) signalling pathways. Conclusions Our findings uncover a critical role of the Bmal1-Junb-AKT/ERK axis in the circadian control of ConA-induced hepatitis and provide new insights into the prevention and treatment of AIH. Impact and Implications This study unveils a critical role of the Bmal1-Junb-AKT/ERK axis in the circadian control of ConA-induced liver injury, providing new insights into the prevention and treatment of immune-mediated hepatitis, including autoimmune hepatitis (AIH). The findings have scientific implications as they enhance our understanding of the circadian regulation of immune responses in liver diseases. Furthermore, clinically, this research offers opportunities for optimising treatment strategies in immune-mediated hepatitis by considering the timing of therapeutic interventions.
Collapse
Affiliation(s)
- Zhaiyi Liu
- School of Bioengineering, Dalian University of Technology, Dalian, China
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Jiayang Zhang
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, China
| | - Shuyao Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hui Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Baoyin Ren
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Jiazhi Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhiyue Bao
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Jiaxin Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Meina Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
4
|
Antonova DV, Gnatenko DA, Kotova ES, Pleshkan VV, Kuzmich AI, Didych DA, Sverdlov ED, Alekseenko IV. Cell-specific expression of the FAP gene is regulated by enhancer elements. Front Mol Biosci 2023; 10:1111511. [PMID: 36825204 PMCID: PMC9941708 DOI: 10.3389/fmolb.2023.1111511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
Fibroblast activation protein (FAP) is an integral membrane serine protease that acts as both dipeptidyl peptidase and collagenase. In recent years, FAP has attracted considerable attention due to its specific upregulation in multiple types of tumor cell populations, including cancer cells in various cancer types, making FAP a potential target for therapy. However, relatively few papers pay attention to the mechanisms driving the cell-specific expression of the FAP gene. We found no correlation between the activities of the two FAP promoter variants (short and long) and the endogenous FAP mRNA expression level in several cell lines with different FAP expression levels. This suggested that other mechanisms may be responsible for specific transcriptional regulation of the FAP gene. We analyzed the distribution of known epigenetic and structural chromatin marks in FAP-positive and FAP-negative cell lines and identified two potential enhancer-like elements (E1 and E2) in the FAP gene locus. We confirmed the specific enrichment of H3K27ac in the putative enhancer regions in FAP-expressing cells. Both the elements exhibited enhancer activity independently of each other in the functional test by increasing the activity of the FAP promoter variants to a greater extent in FAP-expressing cell lines than in FAP-negative cell lines. The transcription factors AP-1, CEBPB, and STAT3 may be involved in FAP activation in the tumors. We hypothesized the existence of a positive feedback loop between FAP and STAT3, which may have implications for developing new approaches in cancer therapy.
Collapse
Affiliation(s)
- Dina V. Antonova
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Department of Genomics and Postgenomic Technologies, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry A. Gnatenko
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Department of Genomics and Postgenomic Technologies, Russian Academy of Sciences, Moscow, Russia
| | - Elena S. Kotova
- Laboratory of Human Molecular Genetics, FSBI Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, Russia
| | - Victor V. Pleshkan
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Department of Genomics and Postgenomic Technologies, Russian Academy of Sciences, Moscow, Russia,Gene Oncotherapy Sector, Institute of Molecular Genetics, National Research Centre “Kurchatov Institute”, Moscow, Russia
| | - Alexey I. Kuzmich
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Department of Genomics and Postgenomic Technologies, Russian Academy of Sciences, Moscow, Russia,Gene Oncotherapy Sector, Institute of Molecular Genetics, National Research Centre “Kurchatov Institute”, Moscow, Russia
| | - Dmitry A. Didych
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Department of Genomics and Postgenomic Technologies, Russian Academy of Sciences, Moscow, Russia,*Correspondence: Dmitry A. Didych,
| | - Eugene D. Sverdlov
- Kurchatov Center for Genome Research, National Research Centre “Kurchatov Institute”, Moscow, Russia
| | - Irina V. Alekseenko
- Gene Immunooncotherapy Group, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Department of Genomics and Postgenomic Technologies, Russian Academy of Sciences, Moscow, Russia,Gene Oncotherapy Sector, Institute of Molecular Genetics, National Research Centre “Kurchatov Institute”, Moscow, Russia,Laboratory of Epigenetics, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov, Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
5
|
Bronisz E, Cudna A, Wierzbicka A, Kurkowska-Jastrzębska I. Blood-Brain Barrier-Associated Proteins Are Elevated in Serum of Epilepsy Patients. Cells 2023; 12:cells12030368. [PMID: 36766708 PMCID: PMC9913812 DOI: 10.3390/cells12030368] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/08/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Blood-brain barrier (BBB) dysfunction emerges as one of the mechanisms underlying the induction of seizures and epileptogenesis. There is growing evidence that seizures also affect BBB, yet only scarce data is available regarding serum levels of BBB-associated proteins in chronic epilepsy. In this study, we aimed to assess serum levels of molecules associated with BBB in patients with epilepsy in the interictal period. Serum levels of MMP-9, MMP-2, TIMP-1, TIMP-2, S100B, CCL-2, ICAM-1, P-selectin, and TSP-2 were examined in a group of 100 patients who were seizure-free for a minimum of seven days and analyzed by ELISA. The results were compared with an age- and sex-matched control group. Serum levels of MMP-9, MMP-2, TIMP-1, TIMP-2 and S100B were higher in patients with epilepsy in comparison to control group (p < 0.0001; <0.0001; 0.001; <0.0001; <0.0001, respectively). Levels of CCL-2, ICAM-1, P-selectin and TSP-2 did not differ between the two groups. Serum levels of MMP-9, MMP-2, TIMP-1, TIMP-2 and S100B are elevated in patients with epilepsy in the interictal period, which suggests chronic processes of BBB disruption and restoration. The pathological process initiating epilepsy, in addition to seizures, is probably the factor contributing to the elevation of serum levels of the examined molecules.
Collapse
Affiliation(s)
- Elżbieta Bronisz
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
- Correspondence:
| | - Agnieszka Cudna
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Aleksandra Wierzbicka
- Sleep Disorders Center, Department of Clinical Neurophysiology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Iwona Kurkowska-Jastrzębska
- Sleep Disorders Center, Department of Clinical Neurophysiology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| |
Collapse
|
6
|
Bronisz E, Cudna A, Wierzbicka A, Kurkowska-Jastrzębska I. Serum Proteins Associated with Blood-Brain Barrier as Potential Biomarkers for Seizure Prediction. Int J Mol Sci 2022; 23:ijms232314712. [PMID: 36499038 PMCID: PMC9740683 DOI: 10.3390/ijms232314712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
As 30% of epileptic patients remain drug-resistant, seizure prediction is vital. Induction of epileptic seizure is a complex process that can depend on factors such as intrinsic neuronal excitability, changes in extracellular ion concentration, glial cell activity, presence of inflammation and activation of the blood−brain barrier (BBB). In this study, we aimed to assess if levels of serum proteins associated with BBB can predict seizures. Serum levels of MMP-9, MMP-2, TIMP-1, TIMP-2, S100B, CCL-2, ICAM-1, P-selectin, and TSP-2 were examined in a group of 49 patients with epilepsy who were seizure-free for a minimum of seven days and measured by ELISA. The examination was repeated after 12 months. An extensive medical history was taken, and patients were subjected to a follow-up, including a detailed history of seizures. Serum levels of MMP-2, MMP-9, TIMP-1, CCL-2, and P-selectin differed between the two time points (p < 0.0001, p < 0.0001, p < 0.0001, p < 0.0001, p = 0.0035, respectively). General linear model analyses determined the predictors of seizures. Levels of MMP-2, MMP-9, and CCL-2 were found to influence seizure count in 1, 3, 6, and 12 months of observation. Serum levels of MMP-2, MMP-9, and CCL-2 may be considered potential biomarkers for seizure prediction and may indicate BBB activation.
Collapse
Affiliation(s)
- Elżbieta Bronisz
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
- Correspondence:
| | - Agnieszka Cudna
- Second Department of Neurology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | - Aleksandra Wierzbicka
- Sleep Disorders Center, Department of Clinical Neurophysiology, Institute of Psychiatry and Neurology, 02-957 Warsaw, Poland
| | | |
Collapse
|
7
|
Gross C, Le-Bel G, Desjardins P, Benhassine M, Germain L, Guérin SL. Contribution of the Transcription Factors Sp1/Sp3 and AP-1 to Clusterin Gene Expression during Corneal Wound Healing of Tissue-Engineered Human Corneas. Int J Mol Sci 2021; 22:12426. [PMID: 34830308 PMCID: PMC8621254 DOI: 10.3390/ijms222212426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/16/2022] Open
Abstract
In order to reduce the need for donor corneas, understanding of corneal wound healing and development of an entirely tissue-engineered human cornea (hTECs) is of prime importance. In this study, we exploited the hTEC to determine how deep wound healing affects the transcriptional pattern of corneal epithelial cells through microarray analyses. We demonstrated that the gene encoding clusterin (CLU) has its expression dramatically repressed during closure of hTEC wounds. Western blot analyses confirmed a strong reduction in the expression of the clusterin isoforms after corneal damage and suggest that repression of CLU gene expression might be a prerequisite to hTEC wound closure. Transfection with segments from the human CLU gene promoter revealed the presence of three regulatory regions: a basal promoter and two more distal negative regulatory regions. The basal promoter bears DNA binding sites for very potent transcription factors (TFs): Activator Protein-1 (AP-1) and Specificity protein-1 and 3 (Sp1/Sp3). By exploiting electrophoretic mobility shift assays (EMSA), we demonstrated that AP-1 and Sp1/Sp3 have their DNA binding site overlapping with one another in the basal promoter of the CLU gene in hCECs. Interestingly, expression of both these TFs is reduced (at the protein level) during hTEC wound healing, thereby contributing to the extinction of CLU gene expression during that process. The results of this study contribute to a better understanding of the molecular mechanisms accounting for the repression of CLU gene expression during corneal wound healing.
Collapse
Affiliation(s)
- Christelle Gross
- Centre Universitaire d’Ophtalmologie-Recherche (CUO-Recherche), Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada; (C.G.); (G.L.-B.); (P.D.); (M.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Gaëtan Le-Bel
- Centre Universitaire d’Ophtalmologie-Recherche (CUO-Recherche), Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada; (C.G.); (G.L.-B.); (P.D.); (M.B.); (L.G.)
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Pascale Desjardins
- Centre Universitaire d’Ophtalmologie-Recherche (CUO-Recherche), Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada; (C.G.); (G.L.-B.); (P.D.); (M.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Manel Benhassine
- Centre Universitaire d’Ophtalmologie-Recherche (CUO-Recherche), Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada; (C.G.); (G.L.-B.); (P.D.); (M.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Lucie Germain
- Centre Universitaire d’Ophtalmologie-Recherche (CUO-Recherche), Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada; (C.G.); (G.L.-B.); (P.D.); (M.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Sylvain L. Guérin
- Centre Universitaire d’Ophtalmologie-Recherche (CUO-Recherche), Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada; (C.G.); (G.L.-B.); (P.D.); (M.B.); (L.G.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada
- Département d’Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
8
|
Wutschka J, Kast B, Sator-Schmitt M, Appak-Baskoy S, Hess J, Sinn HP, Angel P, Schorpp-Kistner M. JUNB suppresses distant metastasis by influencing the initial metastatic stage. Clin Exp Metastasis 2021; 38:411-423. [PMID: 34282521 PMCID: PMC8318945 DOI: 10.1007/s10585-021-10108-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/23/2021] [Indexed: 01/01/2023]
Abstract
The complex interactions between cells of the tumor microenvironment and cancer cells are considered a major determinant of cancer progression and metastasis. Yet, our understanding of the mechanisms of metastatic disease is not sufficient to successfully treat patients with advanced-stage cancer. JUNB is a member of the AP-1 transcription factor family shown to be frequently deregulated in human cancer and associated with invasion and metastasis. A strikingly high stromal JUNB expression in human breast cancer samples prompted us to functionally investigate the consequences of JUNB loss in cells of the tumor microenvironment on cancer progression and metastasis in mice. To adequately mimic the clinical situation, we applied a syngeneic spontaneous breast cancer metastasis model followed by primary tumor resection and identified stromal JUNB as a potent suppressor of distant metastasis. Comprehensive characterization of the JUNB-deficient tumor microenvironment revealed a strong influx of myeloid cells into primary breast tumors and lungs at early metastatic stage. In these infiltrating neutrophils, BV8 and MMP9, proteins promoting angiogenesis and tissue remodeling, were specifically upregulated in a JUNB-dependent manner. Taken together, we established stromal JUNB as a strong suppressor of distant metastasis. Consequently, therapeutic strategies targeting AP-1 should be carefully designed not to interfere with stromal JUNB expression as this may be detrimental for cancer patients.
Collapse
Affiliation(s)
- Juliane Wutschka
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Faculty of Biosciences, University Heidelberg, Heidelberg, Germany
| | - Bettina Kast
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Melanie Sator-Schmitt
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Sila Appak-Baskoy
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Chemistry and Biology, Ryerson University, Toronto, ON, Canada
- iBEST (Institute of Biomedical Engineering, Science and Technology), Toronto, ON, Canada
| | - Jochen Hess
- Department of Otorhinolaryngology, Head and Neck Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Research Group Molecular Mechanisms of Head and Neck Tumors, DKFZ, Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Marina Schorpp-Kistner
- Division of Signal Transduction and Growth Control, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
9
|
Ganguly K, Trigun SK. Mapping Connectome in Mammalian Brain: A Novel Approach by Bioengineering Neuro-Glia specific Vectors. J Theor Biol 2020; 496:110244. [PMID: 32171712 DOI: 10.1016/j.jtbi.2020.110244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 10/24/2022]
Abstract
The connectome is the comprehensive map of the brain represented by wiring diagram of the full set of neuro-glia and synapses within entire brain of an organism. Some recent scientific efforts have successfully been made to visualize such map at neuro-glial networking level, however, capturing it as one unit of the entire brain have never been elucidated. Moreover, in order to derive structure-function relationship of different brain regions in response to a defined stimulus, there is a need to elucidate the connectome at single neuro-glial ensemble level after brain is challenged with the known memory function. This needs developing molecular approaches to tag neuro-glial activities in response to a conditioned brain function. Such approaches of using specific molecular tags have been tried to visualize independently neuron and glial specific events in response to a memory function, however, they could not tag the connectome together at single neuro-glia ensemble level. Therefore, there is a need to develop new methods for mapping entire connectome up to a single neuro-glial precision and resolution, with a purpose of tagging specific brain region accountable to execute a special memory formation process. The present hypothetical paper aims to propose a novel molecular method to generate the structural connectome at neuro-glial level in mice brain. Herein, we propose to tag the entire connectome at neuro-glia precision by generating a transgenic mice via transposing and recombining engineered novel "Neuro-Glia specific Vectors" (NGVs: specific to excitatory neurons, inhibitory neurons and glial cells) vis a vis "Transcriptional/ Translational Messenger (TMs: specific to metalloproteinases, MMP-9) coupled with different color protein tags, followed by the Clarity. Herein, the NGVs will be translated via Neuro-glia specific promoters, while TMs will be translated via endogenous MMP-9 promoter in all neuro-glial cells. The viability of all constructs will be verified in cortical/ hippocampal culture by inducing them to undergo chemically induced long term potentionation (cLTP) following visualization of different colored pattern. This will be further confirmed by Immunostaning, Western Blot and RT-PCR analysis. Additionally, in this approach, one can decipher the dynamics of molecular and cellular events associated with MMP-9 seretome by monitoring the trafficking of tagged endogenous MMP-9 protein after neuronal stimulation by cLTP in vitro. However, for visualizing complete connectome, the adult transgenic mice will be challenged with fear consolidation (Fear context and contextual cue) tests followed by Clarity coupled Light Sheet Microscopy to analyze neuro-glia ensemble following whole brain imaging.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005 Uttar Pradesh, India
| | - Surendra Kumar Trigun
- Biochemistry and Molecular Biology Unit, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221005 Uttar Pradesh, India.
| |
Collapse
|
10
|
Sun ZQ, Meng FH, Tu LX, Sun L. Myricetin attenuates the severity of seizures and neuroapoptosis in pentylenetetrazole kindled mice by regulating the of BDNF-TrkB signaling pathway and modulating matrix metalloproteinase-9 and GABA A. Exp Ther Med 2019; 17:3083-3091. [PMID: 30906480 PMCID: PMC6425265 DOI: 10.3892/etm.2019.7282] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 10/19/2018] [Indexed: 12/21/2022] Open
Abstract
Currently available antiepileptic drugs are effective; however, frequently associated with adverse effects that limit their therapeutic value. Compounds that target the molecular events underlying epilepsy, with minor or no adverse effects, would be of clinical value. Matrix metalloproteinase-9 (MMP-9) and the brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling pathway may be involved in epileptogenesis. The current study investigated the effects of the plant-derived hydroxyflavone, myricetin, in a pentylenetetrazole (PTZ)-induced mouse model of epilepsy. Mice received an intraperitoneal injection of 35 mg/kg body weight PTZ on alternate days (13 injections) and were observed for 30 min following each PTZ injection. Myricetin (100 or 200 mg/kg body weight) was administered orally to the treatment groups (n=18/group) for 26 days, 30 min prior to each PTZ injection. Treatment with myricetin reduced seizure and mortality rates. Increased apoptotic cell count and elevated expression levels of apoptotic proteins caused by PTZ kindling were downregulated following treatment with myricetin. The BDNF-TrkB signaling pathway and MMP-9 expression levels were regulated by myricetin. Expression of γ-aminobutyric acid A (GABA) receptor and glutamic acid decarboxylase 65, as well as the glutamate/GABA balance, were restored following treatment with myricetin. The results of the present study indicated that myricetin may exert protective effects by regulating the molecular events associated with epileptogenesis.
Collapse
Affiliation(s)
- Zhi-Qing Sun
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Fan-Hua Meng
- Department of Neurology, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Li-Xiang Tu
- Department of Infectious Disease, Linyi People's Hospital, Linyi, Shandong 276003, P.R. China
| | - Lei Sun
- Department of Emergency, Hedong People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
11
|
Zybura-Broda K, Wolder-Gontarek M, Ambrozek-Latecka M, Choros A, Bogusz A, Wilemska-Dziaduszycka J, Rylski M. HuR (Elavl1) and HuB (Elavl2) Stabilize Matrix Metalloproteinase-9 mRNA During Seizure-Induced Mmp-9 Expression in Neurons. Front Neurosci 2018; 12:224. [PMID: 29686606 PMCID: PMC5900018 DOI: 10.3389/fnins.2018.00224] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/22/2018] [Indexed: 01/28/2023] Open
Abstract
Matrix metalloproteinase-9 (Mmp-9) is involved in different general and cell-type–specific processes, both in neuronal and non-neuronal cells. Moreover, it is implicated in an induction or progression of various human disorders, including diseases of the central nervous system. Mechanisms regulating activity-driven Mmp-9 expression in neurons are still not fully understood. Here, we show that stabilization of Mmp-9 mRNA is one of the factors responsible for the neuronal activity-evoked upregulation of Mmp-9 mRNA expression in hippocampal neurons. Furthermore, we demonstrate that the molecular mechanism related to this stabilization is dependent on the neuronal seizure-triggered transiently increased binding of the mRNA stability-inducing protein, HuR, to ARE1 and ARE4 motifs of the 3′UTR for Mmp-9 mRNA as well as the stably augmented association of another mRNA-stabilizing protein, HuB, to the ARE1 element of the 3′UTR. Intriguingly, we demonstrate further that both HuR and HuB are crucial for an incidence of Mmp-9 mRNA stabilization after neuronal activation. This study identifies Mmp-9 mRNA as the first HuB target regulated by mRNA stabilization in neurons. Moreover, these results are the first to describe an existence of HuR-dependent mRNA stabilization in neurons of the brain.
Collapse
Affiliation(s)
- Katarzyna Zybura-Broda
- Department of Clinical Cytology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | | | | - Artur Choros
- Department of Clinical Cytology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Agnieszka Bogusz
- Department of Clinical Cytology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | | - Marcin Rylski
- Department of Clinical Cytology, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
12
|
Jaworski J, Kalita K, Knapska E. c-Fos and neuronal plasticity: the aftermath of Kaczmarek’s theory. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
13
|
Matrix Metalloproteinase 9 in Epilepsy: The Role of Neuroinflammation in Seizure Development. Mediators Inflamm 2016; 2016:7369020. [PMID: 28104930 PMCID: PMC5220508 DOI: 10.1155/2016/7369020] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/27/2016] [Indexed: 12/11/2022] Open
Abstract
Matrix metalloproteinase 9 is a proteolytic enzyme which is recently one of the more often studied biomarkers. Its possible use as a biomarker of neuronal damage in stroke, heart diseases, tumors, multiple sclerosis, and epilepsy is being widely indicated. In epilepsy, MMP-9 is suggested to play a role in epileptic focus formation and in the stimulation of seizures. The increase of MMP-9 activity in the epileptic focus was observed both in animal models and in clinical studies. MMP-9 contributes to formation of epileptic focus, for example, by remodeling of synapses. Its proteolytic action on the elements of blood-brain barrier and activation of chemotactic processes facilitates accumulation of inflammatory cells and induces seizures. Also modification of glutamatergic transmission by MMP-9 is associated with seizures. In this review we will try to recapitulate the results of previous studies about MMP-9 in terms of its association with epilepsy. We will discuss the mechanisms of its actions and present the results revealed in animal models and clinical studies. We will also provide a comparison of the results of various studies on MMP-9 levels in the context of its possible use as a biomarker of the activity of epilepsy.
Collapse
|
14
|
The multifaceted role of metalloproteinases in physiological and pathological conditions in embryonic and adult brains. Prog Neurobiol 2016; 155:36-56. [PMID: 27530222 DOI: 10.1016/j.pneurobio.2016.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/10/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a large family of ubiquitous extracellular endopeptidases, which play important roles in a variety of physiological and pathological conditions, from the embryonic stages throughout adult life. Their extraordinary physiological "success" is due to concomitant broad substrate specificities and strict regulation of their expression, activation and inhibition levels. In recent years, MMPs have gained increasing attention as significant effectors in various aspects of central nervous system (CNS) physiology. Most importantly, they have been recognized as main players in a variety of brain disorders having different etiologies and evolution. A common aspect of these pathologies is the development of acute or chronic neuroinflammation. MMPs play an integral part in determining the result of neuroinflammation, in some cases turning its beneficial outcome into a harmful one. This review summarizes the most relevant studies concerning the physiology of MMPs, highlighting their involvement in both the developing and mature CNS, in long-lasting and acute brain diseases and, finally, in nervous system repair. Recently, a concerted effort has been made in identifying therapeutic strategies for major brain diseases by targeting MMP activities. However, from this revision of the literature appears clear that MMPs have multifaceted functional characteristics, which modulate physiological processes in multiple ways and with multiple consequences. Therefore, when choosing MMPs as possible targets, great care must be taken to evaluate the delicate balance between their activation and inhibition and to determine at which stage of the disease and at what level they become active in order maximize chances of success.
Collapse
|
15
|
Epigenetics of Epileptogenesis-Evoked Upregulation of Matrix Metalloproteinase-9 in Hippocampus. PLoS One 2016; 11:e0159745. [PMID: 27505431 PMCID: PMC4978505 DOI: 10.1371/journal.pone.0159745] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/07/2016] [Indexed: 01/20/2023] Open
Abstract
Enhanced levels of Matrix Metalloproteinase-9 (MMP-9) have been implicated in the pathogenesis of epilepsy in humans and rodents. Lack of Mmp-9 impoverishes, whereas excess of Mmp-9 facilitates epileptogenesis. Epigenetic mechanisms driving the epileptogenesis-related upregulation of MMP-9 expression are virtually unknown. The aim of this study was to reveal these mechanisms. We analyzed hippocampi extracted from adult and pediatric patients with temporal lobe epilepsy as well as from partially and fully pentylenetetrazole kindled rats. We used a unique approach to the analysis of the kindling model results (inclusion in the analysis of rats being during kindling, and not only a group of fully kindled animals), which allowed us to separate the molecular effects exerted by the epileptogenesis from those related to epilepsy and epileptic activity. Consequently, it allowed for a disclosure of molecular mechanisms underlying causes, and not consequences, of epilepsy. Our data show that the epileptogenesis-evoked upregulation of Mmp-9 expression is regulated by removal from Mmp-9 gene proximal promoter of the two, interweaved potent silencing mechanisms–DNA methylation and Polycomb Repressive Complex 2 (PRC2)-related repression. Demethylation depends on a gradual dissociation of the DNA methyltransferases, Dnmt3a and Dnmt3b, and on progressive association of the DNA demethylation promoting protein Gadd45β to Mmp-9 proximal gene promoter in vivo. The PRC2-related mechanism relies on dissociation of the repressive transcription factor YY1 and the dissipation of the PRC2-evoked trimethylation on Lys27 of the histone H3 from the proximal Mmp-9 promoter chromatin in vivo. Moreover, we show that the DNA hydroxymethylation, a new epigenetic DNA modification, which is localized predominantly in the gene promoters and is particularly abundant in the brain, is not involved in a regulation of MMP-9 expression during the epileptogenesis in the rat hippocampus as well as in the hippocampi of pediatric and adult epileptic patients. Additionally, we have also found that despite of its transient nature, the histone modification H3S10ph is strongly and gradually accumulated during epileptogenesis in the cell nuclei and in the proximal Mmp-9 gene promoter in the hippocampus, which suggests that H3S10ph can be involved in DNA demethylation in mammals, and not only in Neurospora. The study identifies MMP-9 as the first protein coding gene which expression is regulated by DNA methylation in human epilepsy. We present a detailed epigenetic model of the epileptogenesis-evoked upregulation of MMP-9 expression in the hippocampus. To our knowledge, it is the most complex and most detailed mechanism of epigenetic regulation of gene expression ever revealed for a particular gene in epileptogenesis. Our results also suggest for the first time that dysregulation of DNA methylation found in epilepsy is a cause rather than a consequence of this condition.
Collapse
|
16
|
Radwanska K, Schenatto-Pereira G, Ziółkowska M, Łukasiewicz K, Giese KP. Mapping fear memory consolidation and extinction-specific expression of JunB. Neurobiol Learn Mem 2015; 125:106-12. [PMID: 26318493 DOI: 10.1016/j.nlm.2015.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/16/2015] [Accepted: 08/12/2015] [Indexed: 02/06/2023]
Abstract
Understanding the molecular and cellular process specifically regulated during fear memory consolidation and extinction is a critical step toward development of new strategies in the treatment of human fear disorders. Here we used inhibitory component of AP-1 transcription factor, JunB, in order to map brain regions where JunB-dependent transcription is regulated during consolidation and extinction of contextual fear memory. We found that contextual fear memory consolidation induced JunB expression in the medial nucleus and intercalated cells of the amygdala while extinction training induced JunB in the CA1 and CA3 areas of the dorsal hippocampus. JunB upregulation induced by contextual fear memory extinction was absent in alphaCaMKII autophosphorylation-deficient mice which have impaired contextual fear memory extinction. Thus, our data suggest that JunB expression in the medial nucleus and intercalated cells of the amygdala is involved in fear memory consolidation while alphaCaMKII-autophosphorylation-dependent JunB expression in the areas CA1 and CA3 of the dorsal hippocampus regulates fear memory extinction.
Collapse
Affiliation(s)
- Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, Nencki Institute, ul. L. Pasteura 3, Warsaw, Poland; Centre for the Cellular Basis of Behavior, MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 8AF, UK.
| | - Grace Schenatto-Pereira
- Centre for the Cellular Basis of Behavior, MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 8AF, UK; Núcleo de Neurociências (NNC), Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
| | - Magdalena Ziółkowska
- Laboratory of Molecular Basis of Behavior, Nencki Institute, ul. L. Pasteura 3, Warsaw, Poland
| | - Kacper Łukasiewicz
- Laboratory of Molecular Basis of Behavior, Nencki Institute, ul. L. Pasteura 3, Warsaw, Poland
| | - K Peter Giese
- Centre for the Cellular Basis of Behavior, MRC Centre for Neurodegeneration Research, Institute of Psychiatry, King's College London, James Black Centre, 125 Coldharbour Lane, London SE5 8AF, UK
| |
Collapse
|
17
|
Conant K, Allen M, Lim ST. Activity dependent CAM cleavage and neurotransmission. Front Cell Neurosci 2015; 9:305. [PMID: 26321910 PMCID: PMC4531370 DOI: 10.3389/fncel.2015.00305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/27/2015] [Indexed: 12/13/2022] Open
Abstract
Spatially localized proteolysis represents an elegant means by which neuronal activity dependent changes in synaptic structure, and thus experience dependent learning and memory, can be achieved. In vitro and in vivo studies suggest that matrix metalloproteinase and adamalysin activity is concentrated at the cell surface, and emerging evidence suggests that increased peri-synaptic expression, release and/or activation of these proteinases occurs with enhanced excitatory neurotransmission. Synaptically expressed cell adhesion molecules (CAMs) could therefore represent important targets for neuronal activity-dependent proteolysis. Several CAM subtypes are expressed at the synapse, and their cleavage can influence the efficacy of synaptic transmission through a variety of non-mutually exclusive mechanisms. In the following review, we discuss mechanisms that regulate neuronal activity-dependent synaptic CAM shedding, including those that may be calcium dependent. We also highlight CAM targets of activity-dependent proteolysis including neuroligin and intercellular adhesion molecule-5 (ICAM-5). We include discussion focused on potential consequences of synaptic CAM shedding, with an emphasis on interactions between soluble CAM cleavage products and specific pre- and post-synaptic receptors.
Collapse
Affiliation(s)
- Katherine Conant
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Megan Allen
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| | - Seung T Lim
- Department of Neuroscience and Interdisciplinary Program in Neuroscience, Georgetown University Medical Center Washington, DC, USA
| |
Collapse
|
18
|
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: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
19
|
Stawarski M, Stefaniuk M, Wlodarczyk J. Matrix metalloproteinase-9 involvement in the structural plasticity of dendritic spines. Front Neuroanat 2014; 8:68. [PMID: 25071472 PMCID: PMC4091410 DOI: 10.3389/fnana.2014.00068] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/25/2014] [Indexed: 01/01/2023] Open
Abstract
Dendritic spines are the locus for excitatory synaptic transmission in the brain and thus play a major role in neuronal plasticity. The ability to alter synaptic connections includes volumetric changes in dendritic spines that are driven by scaffolds created by the extracellular matrix (ECM). Here, we review the effects of the proteolytic activity of ECM proteases in physiological and pathological structural plasticity. We use matrix metalloproteinase-9 (MMP-9) as an example of an ECM modifier that has recently emerged as a key molecule in regulating the morphology and dysmorphology of dendritic spines that underlie synaptic plasticity and neurological disorders, respectively. We summarize the influence of MMP-9 on the dynamic remodeling of the ECM via the cleavage of extracellular substrates. We discuss its role in the formation, modification, and maintenance of dendritic spines in learning and memory. Finally, we review research that implicates MMP-9 in aberrant synaptic plasticity and spine dysmorphology in neurological disorders, with a focus on morphological abnormalities of dendritic protrusions that are associated with epilepsy.
Collapse
Affiliation(s)
- Michal Stawarski
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mazowieckie, Poland
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mzowieckie, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Mazowieckie, Poland
| |
Collapse
|
20
|
Yoon SS, Jo SA. Mechanisms of Amyloid-β Peptide Clearance: Potential Therapeutic Targets for Alzheimer's Disease. Biomol Ther (Seoul) 2014; 20:245-55. [PMID: 24130920 PMCID: PMC3794520 DOI: 10.4062/biomolther.2012.20.3.245] [Citation(s) in RCA: 137] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 05/01/2012] [Accepted: 05/01/2012] [Indexed: 11/30/2022] Open
Abstract
Amyloid-β peptide (Aβ) is still best known as a molecule to cause Alzheimer’s disease (AD) through accumulation and deposition within the frontal cortex and hippocampus in the brain. Thus, strategies on developing AD drugs have been focused on the reduc-tion of Aβ in the brain. Since accumulation of Aβ depends on the rate of its synthesis and clearance, the metabolic pathway of Aβ in the brain and the whole body should be carefully explored for AD research. Although the synthetic pathway of Aβ is equally important, we summarize primarily the clearance pathway in this paper because the former has been extensively reviewed in previous studies. The clearance of Aβ from the brain is accomplished by several mechanisms which include non-enzymatic and enzymatic pathways. Nonenzymatic pathway includes interstitial fluid drainage, uptake by microglial phagocytosis, and transport across the blood vessel walls into the circulation. Multiple Aβ-degrading enzymes (ADE) implicated in the clearance process have been identified, which include neprilysin, insulin-degrading enzyme, matrix metalloproteinase-9, glutamate carboxypeptidase II and others. A series of studies on Aβ clearance mechanism provide new insight into the pathogenesis of AD at the molecular level and suggest a new target for the development of novel therapeutics.
Collapse
Affiliation(s)
- Sang-Sun Yoon
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 330-714, Republic of Korea
| | | |
Collapse
|
21
|
Tsilibary E, Tzinia A, Radenovic L, Stamenkovic V, Lebitko T, Mucha M, Pawlak R, Frischknecht R, Kaczmarek L. Neural ECM proteases in learning and synaptic plasticity. PROGRESS IN BRAIN RESEARCH 2014; 214:135-57. [PMID: 25410356 DOI: 10.1016/b978-0-444-63486-3.00006-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recent studies implicate extracellular proteases in synaptic plasticity, learning, and memory. The data are especially strong for such serine proteases as thrombin, tissue plasminogen activator, neurotrypsin, and neuropsin as well as matrix metalloproteinases, MMP-9 in particular. The role of those enzymes in the aforementioned phenomena is supported by the experimental results on the expression patterns (at the gene expression and protein and enzymatic activity levels) and functional studies, including knockout mice, specific inhibitors, etc. Counterintuitively, the studies have shown that the extracellular proteolysis is not responsible mainly for an overall degradation of the extracellular matrix (ECM) and loosening perisynaptic structures, but rather allows for releasing signaling molecules from the ECM, transsynaptic proteins, and latent form of growth factors. Notably, there are also indications implying those enzymes in the major neuropsychiatric disorders, probably by contributing to synaptic aberrations underlying such diseases as schizophrenia, bipolar, autism spectrum disorders, and drug addiction.
Collapse
Affiliation(s)
- Effie Tsilibary
- Institute of Biosciences and Applications, NCSR "Demokritos", Athens, Greece
| | - Athina Tzinia
- Institute of Biosciences and Applications, NCSR "Demokritos", Athens, Greece
| | - Lidija Radenovic
- Center for Laser Microscopy, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Vera Stamenkovic
- Center for Laser Microscopy, Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Tomasz Lebitko
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Warsaw, Poland
| | | | | | - Renato Frischknecht
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Leszek Kaczmarek
- Department of Molecular and Cellular Neurobiology, Nencki Institute, Warsaw, Poland.
| |
Collapse
|
22
|
Reward learning requires activity of matrix metalloproteinase-9 in the central amygdala. J Neurosci 2013; 33:14591-600. [PMID: 24005309 DOI: 10.1523/jneurosci.5239-12.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Learning how to avoid danger and pursue reward depends on negative emotions motivating aversive learning and positive emotions motivating appetitive learning. The amygdala is a key component of the brain emotional system; however, an understanding of how various emotions are differentially processed in the amygdala has yet to be achieved. We report that matrix metalloproteinase-9 (MMP-9, extracellularly operating enzyme) in the central nucleus of the amygdala (CeA) is crucial for appetitive, but not for aversive, learning in mice. The knock-out of MMP-9 impairs appetitively motivated conditioning, but not an aversive one. MMP-9 is present at the excitatory synapses in the CeA with its activity greatly enhanced after the appetitive training. Finally, blocking extracellular MMP-9 activity with its inhibitor TIMP-1 provides evidence that local MMP-9 activity in the CeA is crucial for the appetitive, but not for aversive, learning.
Collapse
|
23
|
Mizoguchi H, Yamada K. Roles of matrix metalloproteinases and their targets in epileptogenesis and seizures. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2013; 11:45-52. [PMID: 24023547 PMCID: PMC3766754 DOI: 10.9758/cpn.2013.11.2.45] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 04/05/2013] [Accepted: 04/19/2013] [Indexed: 01/01/2023]
Abstract
Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) remodel the pericellular environment by regulating the cleavage of extracellular matrix proteins, cell surface components, neurotransmitter receptors, and growth factors, which together regulate cell adhesion, synaptogenesis, synaptic plasticity, and long-term potentiation. Increased MMP activity and dysregulation of the balance between MMPs and TIMPs have also been implicated in various pathological conditions. Recent studies have suggested that prolonged seizures are associated with high MMP levels in serum and neural tissues, and certain extracellular macromolecule targets may influence the pathogenesis of epilepsy and seizure. In this review, we discuss the roles of MMP activation in animal models of epilepsy.
Collapse
Affiliation(s)
- Hiroyuki Mizoguchi
- Futuristic Environmental Simulation Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | | |
Collapse
|
24
|
Ganguly K, Rejmak E, Mikosz M, Nikolaev E, Knapska E, Kaczmarek L. Matrix metalloproteinase (MMP) 9 transcription in mouse brain induced by fear learning. J Biol Chem 2013; 288:20978-20991. [PMID: 23720741 PMCID: PMC3774367 DOI: 10.1074/jbc.m113.457903] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 05/24/2013] [Indexed: 12/18/2022] Open
Abstract
Memory formation requires learning-based molecular and structural changes in neurons, whereas matrix metalloproteinase (MMP) 9 is involved in the synaptic plasticity by cleaving extracellular matrix proteins and, thus, is associated with learning processes in the mammalian brain. Because the mechanisms of MMP-9 transcription in the brain are poorly understood, this study aimed to elucidate regulation of MMP-9 gene expression in the mouse brain after fear learning. We show here that contextual fear conditioning markedly increases MMP-9 transcription, followed by enhanced enzymatic levels in the three major brain structures implicated in fear learning, i.e. the amygdala, hippocampus, and prefrontal cortex. To reveal the role of AP-1 transcription factor in MMP-9 gene expression, we have used reporter gene constructs with specifically mutated AP-1 gene promoter sites. The constructs were introduced into the medial prefrontal cortex of neonatal mouse pups by electroporation, and the regulation of MMP-9 transcription was studied after contextual fear conditioning in the adult animals. Specifically, -42/-50- and -478/-486-bp AP-1 binding motifs of the mouse MMP-9 promoter sequence have been found to play a major role in MMP-9 gene activation. Furthermore, increases in MMP-9 gene promoter binding by the AP-1 transcription factor proteins c-Fos and c-Jun have been demonstrated in all three brain structures under investigation. Hence, our results suggest that AP-1 acts as a positive regulator of MMP-9 transcription in the brain following fear learning.
Collapse
Affiliation(s)
| | | | - Marta Mikosz
- Laboratory of Neurobiology of Emotions, Nencki Institute, Pasteur 3, 02-093 Warsaw, Poland
| | | | - Ewelina Knapska
- Laboratory of Neurobiology of Emotions, Nencki Institute, Pasteur 3, 02-093 Warsaw, Poland
| | | |
Collapse
|
25
|
Brain-derived neurotrophic factor induces matrix metalloproteinase 9 expression in neurons via the serum response factor/c-Fos pathway. Mol Cell Biol 2013; 33:2149-62. [PMID: 23508111 DOI: 10.1128/mcb.00008-13] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays a pivotal role in the regulation of the transcription of genes that encode proplasticity proteins. In the present study, we provide evidence that stimulation of rat primary cortical neurons with BDNF upregulates matrix metalloproteinase 9 (MMP-9) mRNA and protein levels and increases enzymatic activity. The BDNF-induced MMP-9 transcription was dependent on extracellular signal-regulated kinase 1/2 (ERK1/2) pathway and c-Fos expression. Overexpression of AP-1 dimers in neurons led to MMP-9 promoter activation, with the most potent being those that contained c-Fos, whereas knockdown of endogenous c-Fos by small hairpin RNA (shRNA) reduced BDNF-mediated MMP-9 transcription. Additionally, mutation of the proximal AP-1 binding site in the MMP-9 promoter inhibited the activation of MMP-9 transcription. BDNF stimulation of neurons induced binding of endogenous c-Fos to the proximal MMP-9 promoter region. Furthermore, as the c-Fos gene is a known target of serum response factor (SRF), we investigated whether SRF contributes to MMP-9 transcription. Inhibition of SRF and its cofactors by either overexpression of dominant negative mutants or shRNA decreased MMP-9 promoter activation. In contrast, MMP-9 transcription was not dependent on CREB activity. Finally, we showed that neuronal activity stimulates MMP-9 transcription in a tyrosine kinase receptor B (TrkB)-dependent manner.
Collapse
|
26
|
Hoehna Y, Uckermann O, Luksch H, Stefovska V, Marzahn J, Theil M, Gorkiewicz T, Gawlak M, Wilczynski GM, Kaczmarek L, Ikonomidou C. Matrix metalloproteinase 9 regulates cell death following pilocarpine-induced seizures in the developing brain. Neurobiol Dis 2012; 48:339-47. [DOI: 10.1016/j.nbd.2012.06.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 06/24/2012] [Accepted: 06/27/2012] [Indexed: 12/28/2022] Open
|
27
|
Abstract
Focal cortical dysplasia (FCD) is a developmental brain disorder characterized by localized abnormalities of cortical layering and neuronal morphology. It is associated with pharmacologically intractable forms of epilepsy in both children and adults. The mechanisms that underlie FCD-associated seizures and lead to the progression of the disease are unclear. Matrix metalloproteinases (MMPs) are enzymes that are able to influence neuronal function through extracellular proteolysis in various normal and pathological conditions. The results of experiments that have used rodent models showed that extracellular MMP-9 can play an important role in epileptogenesis. However, no studies have shown that MMP-9 is involved in the pathogenesis of human epilepsy. The aim of the present study was to determine whether MMP-9 plays a role in intractable epilepsy. Using an unbiased antibody microarray approach, we found that up regulation of MMP-9 is prominent and consistent in FCD tissue derived from epilepsy surgery, regardless of the patient's age. Additionally, an up regulation of MMP-1, -2, -8, -10, and -13 was found but was either less pronounced or limited only to adult cases. In the dysplastic cortex, immunohistochemistry revealed that the highest MMP-9 immuno reactivity occurred in the cytoplasm of abnormal neurons and balloon cells. The neuronal over expression of MMP-9 also occurred in sclerotic hippocampi that were excised together with the dysplastic cortex, but sclerotic hippocampi were free of dysplastic features. In both locations, MMP-9 was also found in reactive astrocytes, albeit to a lesser extent. At the subcellular level, increased MMP-9 immunoreactivity was prominently upregulated at synapses. Thus, although upregulation of the enzyme in FCD is not causally linked to the developmental malformation, it may be a result of ongoing abnormal synaptic plasticity. The present findings support the hypothesis of the pathogenic role of MMP-9 in human epilepsy and may stimulate discussions about whether MMPs could be novel therapeutic targets for intractable epilepsy.
Collapse
|
28
|
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: 209] [Impact Index Per Article: 16.1] [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
|
29
|
Subik K, Shu L, Wu C, Liang Q, Hicks D, Boyce B, Schiffhauer L, Chen D, Chen C, Tang P, Xing L. The ubiquitin E3 ligase WWP1 decreases CXCL12-mediated MDA231 breast cancer cell migration and bone metastasis. Bone 2012; 50:813-23. [PMID: 22266093 PMCID: PMC3439807 DOI: 10.1016/j.bone.2011.12.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 12/27/2011] [Accepted: 12/30/2011] [Indexed: 01/17/2023]
Abstract
Advanced breast cancers preferentially metastasize to bone where cells in the bone microenvironment produce factors that enhance breast cancer cell homing and growth. Expression of the ubiquitin E3 ligase WWP1 is increased in some breast cancers, but its role in bone metastasis has not been investigated. Here, we studied the effects of WWP1 and itch, its closest family member, on breast cancer bone metastasis. First, we immunostained a multi-tumor tissue microarray and a breast cancer tissue microarray and demonstrated that WWP1 and ITCH are expressed in some of breast cancer cases. We then knocked down WWP1 or itch in MDA-MB-231 breast cancer cells using shRNA and inoculated these cells and control cells into the left ventricle of athymic nude mice. Radiographs showed that mice given shWWP1 cells had more osteolytic lesions than mice given control MDA-MB-231 cells. Histologic analysis confirmed osteolysis and showed significantly increased tumor area in bone marrow of the mice. WWP1 knockdown did not affect cell growth, survival or osteoclastogenic potential, but markedly increased cell migration toward a CXCL12 gradient in vitro. Furthermore, WWP1 knockdown significantly reduced CXCL12-induced CXCR4 lysosomal trafficking and degradation. In contrast, itch knockdown had no effect on MDA-MB-231 cell bone metastasis. Taken together, these findings demonstrate that WWP1 negatively regulates cell migration to CXCL12 by limiting CXCR4 degradation to promote breast cancer metastasis to bone and highlight the potential utility of WWP1 as a prognostic indicator for breast cancer bone metastasis.
Collapse
Affiliation(s)
- Kristina Subik
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lei Shu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Chengyu Wu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Qianqian Liang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David Hicks
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brendan Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Linda Schiffhauer
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Di Chen
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Ping Tang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Corresponding author at: Department of Pathology and Laboratory Medicine, 601 Elmwood Ave, Box 626, Rochester, NY 14642, USA. Fax: +1 585 756 4468. (L. Xing)
| |
Collapse
|
30
|
Lee KH, Kim JR. Regulation of HGF-mediated cell proliferation and invasion through NF-κB, JunB, and MMP-9 cascades in stomach cancer cells. Clin Exp Metastasis 2012; 29:263-72. [DOI: 10.1007/s10585-011-9449-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Accepted: 12/27/2011] [Indexed: 11/30/2022]
|
31
|
Matrix metalloproteinases contribute to neuronal dysfunction in animal models of drug dependence, Alzheimer's disease, and epilepsy. Biochem Res Int 2011; 2011:681385. [PMID: 22235372 PMCID: PMC3253438 DOI: 10.1155/2011/681385] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/17/2011] [Indexed: 01/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) remodel the pericellular environment by regulating the cleavage of extracellular matrix proteins, cell surface components, neurotransmitter receptors, and growth factors that mediate cell adhesion, synaptogenesis, synaptic plasticity, and long-term potentiation. Interestingly, increased MMP activity and dysregulation of the balance between MMPs and TIMPs have also been implicated in various pathologic conditions. In this paper, we discuss various animal models that suggest that the activation of the gelatinases MMP-2 and MMP-9 is involved in pathogenesis of drug dependence, Alzheimer's disease, and epilepsy.
Collapse
|
32
|
Matrix metalloproteinase-9 contributes to kindled seizure development in pentylenetetrazole-treated mice by converting pro-BDNF to mature BDNF in the hippocampus. J Neurosci 2011; 31:12963-71. [PMID: 21900575 DOI: 10.1523/jneurosci.3118-11.2011] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Recurrent seizure activity has been shown to induce a variety of permanent structural changes in the brain. Matrix metalloproteinases (MMPs) function to promote neuronal plasticity, primarily through cleavage of extracellular matrix proteins. Here, we investigated the role of MMP-9 in the development of pentylenetetrazole (PTZ)-induced kindled seizure in mice. Repeated treatment with PTZ (40 mg/kg) produced kindled seizure, which was accompanied by enhanced MMP-9 activity and expression in the hippocampus. No change in MMP-9 activity was observed in the hippocampi of mice with generalized tonic seizure following single administration of PTZ (60 mg/kg). MMP-9 colocalized with the neuronal marker NeuN and the glial marker GFAP in the dentate gyrus of the kindled mouse hippocampus. Coadministration of diazepam or MK-801 with PTZ inhibited the development of kindling and the increased MMP-9 levels in the hippocampus. Marked suppression of kindled seizure progression in response to repeated PTZ treatment was observed in MMP-9((-/-)) mice compared with wild-type mice, an observation that was accompanied by decreased hippocampal levels of mature brain-derived neurotrophic factor. Microinjecting the BDNF scavenger TrkB-Fc into the right ventricle before each PTZ treatment significantly suppressed the development of kindling in wild-type mice, whereas no effect was observed in MMP-9((-/-)) mice. On the other hand, bilateral injections of pro-BDNF into the hippocampal dentate gyrus significantly enhanced kindling in wild-type mice but not MMP-9((-/-)) mice. These findings suggest that MMP-9 is involved in the progression of behavioral phenotypes in kindled mice because of conversion of pro-BDNF to mature BDNF in the hippocampus.
Collapse
|
33
|
Extracellular proteases in epilepsy. Epilepsy Res 2011; 96:191-206. [DOI: 10.1016/j.eplepsyres.2011.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/10/2011] [Accepted: 08/03/2011] [Indexed: 11/20/2022]
|
34
|
Metzincin proteases and their inhibitors: foes or friends in nervous system physiology? J Neurosci 2010; 30:15337-57. [PMID: 21084591 DOI: 10.1523/jneurosci.3467-10.2010] [Citation(s) in RCA: 184] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Members of the metzincin family of metalloproteinases have long been considered merely degradative enzymes for extracellular matrix molecules. Recently, however, there has been growing appreciation for these proteinases and their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs), as fine modulators of nervous system physiology and pathology. Present all along the phylogenetic tree, in all neural cell types, from the nucleus to the synapse and in the extracellular space, metalloproteinases exhibit a complex spatiotemporal profile of expression in the nervous parenchyma and at the neurovascular interface. The irreversibility of their proteolytic activity on numerous biofactors (e.g., growth factors, cytokines, receptors, DNA repair enzymes, matrix proteins) is ideally suited to sustain structural changes that are involved in physiological or postlesion remodeling of neural networks, learning consolidation or impairment, neurodegenerative and neuroinflammatory processes, or progression of malignant gliomas. The present review provides a state of the art overview of the involvement of the metzincin/TIMP system in these processes and the prospects of new therapeutic strategies based on the control of metalloproteinase activity.
Collapse
|
35
|
Loesch M, Zhi HY, Hou SW, Qi XM, Li RS, Basir Z, Iftner T, Cuenda A, Chen G. p38gamma MAPK cooperates with c-Jun in trans-activating matrix metalloproteinase 9. J Biol Chem 2010; 285:15149-15158. [PMID: 20231272 DOI: 10.1074/jbc.m110.105429] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitogen-activated protein kinases (MAPKs) regulate gene expression through transcription factors. However, the precise mechanisms in this critical signal event are largely unknown. Here, we show that the transcription factor c-Jun is activated by p38gamma MAPK, and the activated c-Jun then recruits p38gamma as a cofactor into the matrix metalloproteinase 9 (MMP9) promoter to induce its trans-activation and cell invasion. This signaling event was initiated by hyperexpressed p38gamma that led to increased c-Jun synthesis, MMP9 transcription, and MMP9-dependent invasion through p38gamma interacting with c-Jun. p38gamma requires phosphorylation and its C terminus to bind c-Jun, whereas both c-Jun and p38gamma are required for the trans-activation of MMP9. The active p38gamma/c-Jun/MMP9 pathway also exists in human colon cancer, and there is a coupling of increased p38gamma and MMP9 expression in the primary tissues. These results reveal a new paradigm in which a MAPK acts both as an activator and a cofactor of a transcription factor to regulate gene expression leading to an invasive response.
Collapse
Affiliation(s)
- Mathew Loesch
- Departments of Pharmacology and Toxicology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Hui-Ying Zhi
- Departments of Pharmacology and Toxicology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Song-Wang Hou
- Departments of Pharmacology and Toxicology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Xiao-Mei Qi
- Departments of Pharmacology and Toxicology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Rong-Shan Li
- Departments of Pathology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Zainab Basir
- Departments of Pathology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Thomas Iftner
- Section of Experimental Virology, Institute of Medical Virology and Epidemiology of Viral Diseases, University Hospital of Tübingen, Tübingen D-72076, Germany
| | - Ana Cuenda
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid 28049, Spain
| | - Guan Chen
- Departments of Pharmacology and Toxicology, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226; Research Services, the Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee Medical College of Wisconsin, Milwaukee, Wisconsin 53226.
| |
Collapse
|