1
|
Lan YL, Zou S, Chen R. Update on the intriguing roles of AQP4 expression and redistribution in the progression and treatment of glioma. Ann Med 2024; 56:2401111. [PMID: 39247976 PMCID: PMC11385637 DOI: 10.1080/07853890.2024.2401111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 09/10/2024] Open
Abstract
Aquaporin 4 (AQP4) is abundant in the human brain and has an important role in brain homeostasis and diseases. AQP4 expression has been found to be associated with glioma malignancies. However, the complete understanding of the biological processes and curative importance of AQP4 in glioma remains unclear. The impact of AQP4 subcellular mislocalization on glioma progression and the precise mechanisms regarding AQP4 translocation in glioma need further investigation. In this review, we update recent findings about disturbed AQP4 expression in glioma and explore targeting AQP4 to modulate the glioma progression. Thereafter we discuss some possible mechanisms of action of AQP4 translocations in glioma. The present article offers an appropriate introduction to the potential involvement of AQP4 in the emergence and progression of glioma. Both comprehensive research into the mechanisms and systematically intervention studies focusing on AQP4 are essential. By embracing this strategy, we can obtain a new and insightful outlook on managing cancerous glioma. Although the observations summarized in this review should be confirmed with more studies, we believe that they could provide critical information for the design of more focused research that will allow for systematic and definitive evaluation of the role of AQP4 in glioma treatments.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases, Hangzhou, Zhejiang, China
| | - Shuang Zou
- Key Laboratory of Neuropharmacology and Translational Medicine, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| |
Collapse
|
2
|
Yuan Y, Peng W, Lei J, Zhao Y, Zhao B, Li Y, Wang J, Qu Q. AQP4 Endocytosis-Lysosome Degradation Mediated by MMP-9/β-DG Involved in Diabetes Cognitive Impairment. Mol Neurobiol 2024; 61:8438-8453. [PMID: 38512439 DOI: 10.1007/s12035-024-04085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Cognitive impairment is considered to be one of the important comorbidities of diabetes, but the underlying mechanisms are widely unknown. Aquaporin-4 (AQP4) is the most abundant water channel in the central nervous system, which plays a neuroprotective role in various neurological diseases by maintaining the function of glymphatic system and synaptic plasticity. However, whether AQP4 is involved in diabetes-related cognitive impairment remains unknown. β-dystroglycan (β-DG), a key molecule for anchoring AQP4 on the plasma membrane of astrocytes and avoiding its targeting to lysosomes for degradation, can be cleaved by matrix metalloproteinase-9 (MMP-9). β-DG deficiency can cause a decline in AQP4 via regulating its endocytosis. However, whether cleavage of β-DG can affect the expression of AQP4 remains unreported. In this study, we observed that diabetes mice displayed cognitive disorder accompanied by reduction of AQP4 in prefrontal cortex. And we found that bafilomycin A1, a widely used lysosome inhibitor, could reverse the downregulation of AQP4 in diabetes, further demonstrating that the reduction of AQP4 in diabetes is a result of more endocytosis-lysosome degradation. In further experiments, we found diabetes caused the excessive activation of MMP-9/β-DG which leaded to the loss of connection between AQP4 and β-DG, further inducing the endocytosis of AQP4. Moreover, inhibition of MMP-9/β-DG restored the endocytosis-lysosome degradation of AQP4 and partially alleviated cognitive dysfunction in diabetes. Our study sheds new light on the role of AQP4 in diabetes-associated cognitive disorder. And we provide a promising therapeutic target to reverse the endocytosis-lysosome degradation of AQP4 in diabetes, such as MMP-9/β-DG.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Wei Peng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Jingna Lei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Yi Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Beiyu Zhao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China
| | - Yan Li
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Rd, Xi'an, 710061, China.
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
3
|
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
|
4
|
Anderson MJM, Hayward AN, Smiley AT, Shi K, Pawlak MR, Aird EJ, Grant E, Greenberg L, Aihara H, Evans RL, Ulens C, Gordon WR. Molecular basis of proteolytic cleavage regulation by the extracellular matrix receptor dystroglycan. Structure 2024:S0969-2126(24)00362-9. [PMID: 39305901 DOI: 10.1016/j.str.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/13/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024]
Abstract
The dystrophin-glycoprotein-complex (DGC), anchored by the transmembrane protein dystroglycan, functions to mechanically link the extracellular matrix and actin cytoskeleton. Breaking this connection is associated with diseases such as muscular dystrophy, yet cleavage of dystroglycan by matrix-metalloproteinases (MMPs) remains an understudied mechanism to disrupt the DGC. We determined the crystal structure of the membrane-adjacent domain (amino acids 491-722) of E. coli expressed human dystroglycan to understand MMP cleavage regulation. The structural model includes tandem immunoglobulin-like (IGL) and sperm/enterokinase/agrin-like (SEAL) domains, which support proteolysis in diverse receptors to facilitate mechanotransduction, membrane protection, and viral entry. The structure reveals a C-terminal extension that buries the MMP site by packing into a hydrophobic pocket, a unique mechanism of MMP cleavage regulation. We further demonstrate structure-guided and disease-associated mutations disrupt proteolytic regulation using a cell-surface proteolysis assay. Thus disrupted proteolysis is a potentially relevant mechanism for "breaking" the DGC link to contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Michael J M Anderson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Amanda N Hayward
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Adam T Smiley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Matthew R Pawlak
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Eric J Aird
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA; Currently at Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Eva Grant
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Lauren Greenberg
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Robert L Evans
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Christopher Ulens
- Department of Cellular and Molecular Medicine, Karolinksa University Leuven, 3000 Leuven, Belgium
| | - Wendy R Gordon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
5
|
Li J, Jia S, Song Y, Xu W, Lin J. Ginkgolide B can alleviate spinal cord glymphatic system dysfunction and provide neuroprotection in painful diabetic neuropathy rats by inhibiting matrix metalloproteinase-9. Neuropharmacology 2024; 250:109907. [PMID: 38492884 DOI: 10.1016/j.neuropharm.2024.109907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
The glymphatic system plays a crucial role in maintaining optimal central nervous system (CNS) function by facilitating the removal of metabolic wastes. Aquaporin-4 (AQP4) protein, predominantly located on astrocyte end-feet, is a key pathway for metabolic waste excretion. β-Dystroglycan (β-DG) can anchor AQP4 protein to the end-feet membrane of astrocytes and can be cleaved by matrix metalloproteinase (MMP)-9 protein. Studies have demonstrated that hyperglycemia upregulates MMP-9 expression in the nervous system, leading to neuropathic pain. Ginkgolide B (GB) exerts an inhibitory effect on the MMP-9 protein. In this study, we investigated whether inhibition of MMP-9-mediated β-DG cleavage by GB is involved in the regulation of AQP4 polarity within the glymphatic system in painful diabetic neuropathy (PDN) and exerts neuroprotective effects. The PDN model was established by injecting streptozotocin (STZ). Functional changes in the glymphatic system were observed using magnetic resonance imaging (MRI). The paw withdrawal threshold (PWT) was measured to assess mechanical allodynia. The protein expressions of MMP-9, β-DG, and AQP4 were detected by Western blotting and immunofluorescence. Our findings revealed significant decreases in the efficiency of contrast agent clearance within the spinal glymphatic system of the rats, accompanied by decreased PWT, increased MMP-9 protein expression, decreased β-DG protein expression, and loss of AQP4 polarity. Notably, GB treatment demonstrated the capacity to ameliorate spinal cord glymphatic function by modulating AQP4 polarity through MMP-9 inhibition, offering a promising therapeutic avenue for PDN.
Collapse
Affiliation(s)
- Jiang Li
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Shuaiying Jia
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | | | - Wenmei Xu
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Jingyan Lin
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
6
|
Cook M, Stevenson B, Jacobs LA, Leocadio Victoria D, Cisneros B, Hobbs JK, Stewart CL, Winder SJ. The Role of β-Dystroglycan in Nuclear Dynamics. Cells 2024; 13:431. [PMID: 38474395 DOI: 10.3390/cells13050431] [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: 12/20/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Dystroglycan is a ubiquitously expressed heterodimeric cell-surface laminin receptor with roles in cell adhesion, signalling, and membrane stabilisation. More recently, the transmembrane β-subunit of dystroglycan has been shown to localise to both the nuclear envelope and the nucleoplasm. This has led to the hypothesis that dystroglycan may have a structural role at the nuclear envelope analogous to its role at the plasma membrane. The biochemical fraction of myoblast cells clearly supports the presence of dystroglycan in the nucleus. Deletion of the dystroglycan protein by disruption of the DAG1 locus using CRISPR/Cas9 leads to changes in nuclear size but not overall morphology; moreover, the Young's modulus of dystroglycan-deleted nuclei, as determined by atomic force microscopy, is unaltered. Dystroglycan-disrupted myoblasts are also no more susceptible to nuclear stresses including chemical and mechanical, than normal myoblasts. Re-expression of dystroglycan in DAG1-disrupted myoblasts restores nuclear size without affecting other nuclear parameters.
Collapse
Affiliation(s)
- Matthew Cook
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- A*STAR Skin Research Laboratories, Singapore 138648, Singapore
| | - Ben Stevenson
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Laura A Jacobs
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | | | - Bulmaro Cisneros
- Department of Genetics and Molecular Biology, Centro de Investigación y de Estudios Avanzados, Mexico City 07360, Mexico
| | - Jamie K Hobbs
- Department of Physics and Astronomy, University of Sheffield, Sheffield S3 7RH, UK
| | - Colin L Stewart
- A*STAR Skin Research Laboratories, Singapore 138648, Singapore
| | - Steve J Winder
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
7
|
Si X, Dai S, Fang Y, Tang J, Wang Z, Li Y, Song Z, Chen Y, Liu Y, Zhao G, Zhang B, Pu J. Matrix metalloproteinase-9 inhibition prevents aquaporin-4 depolarization-mediated glymphatic dysfunction in Parkinson's disease. J Adv Res 2024; 56:125-136. [PMID: 36940850 PMCID: PMC10834796 DOI: 10.1016/j.jare.2023.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/31/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023] Open
Abstract
INTRODUCTION The glymphatic system offers a perivascular pathway for the clearance of pathological proteins and metabolites to optimize neurological functions. Glymphatic dysfunction plays a pathogenic role in Parkinson's disease (PD); however, the molecular mechanism of glymphatic dysfunction in PD remains elusive. OBJECTIVE To explore whether matrix metalloproteinase-9 (MMP-9)-mediated β-dystroglycan (β-DG) cleavage is involved in the regulation of aquaporin-4 (AQP4) polarity-mediated glymphatic system in PD. METHODS 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD and A53T mice were used in this study. The glymphatic function was evaluated using ex vivo imaging. TGN-020, an AQP4 antagonist, was administered to investigate the role of AQP4 in glymphatic dysfunction in PD. GM6001, an MMP-9 antagonist, was administered to investigate the role of the MMP-9/β-DG pathway in regulating AQP4. The expression and distribution of AQP4, MMP-9, and β-DG were assessed using western blotting, immunofluorescence, and co-immunoprecipitation. The ultrastructure of basement membrane (BM)-astrocyte endfeet was detected using transmission electron microscopy. Rotarod and open-field tests were performed to evaluate motor behavior. RESULTS Perivascular influx and efflux of cerebral spinal fluid tracers were reduced in MPTP-induced PD mice with impaired AQP4 polarization. AQP4 inhibition aggravated reactive astrogliosis, glymphatic drainage restriction, and dopaminergic neuronal loss in MPTP-induced PD mice. MMP-9 and cleaved β-DG were upregulated in both MPTP-induced PD and A53T mice, with reduced polarized localization of β-DG and AQP4 to astrocyte endfeet. MMP-9 inhibition restored BM-astrocyte endfeet-AQP4 integrity and attenuated MPTP-induced metabolic perturbations and dopaminergic neuronal loss. CONCLUSION AQP4 depolarization contributes to glymphatic dysfunction and aggravates PD pathologies, and MMP-9-mediated β-DG cleavage regulates glymphatic function through AQP4 polarization in PD, which may provide novel insights into the pathogenesis of PD.
Collapse
Affiliation(s)
- Xiaoli Si
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China; Department of Neurology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University, School of Medicine, Yiwu, Zhejiang 322000, China
| | - Shaobing Dai
- Department of Anesthesiology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yi Fang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Jiahui Tang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Zhiyun Wang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yaolin Li
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Zhe Song
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Ying Chen
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Yi Liu
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Guohua Zhao
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China; Department of Neurology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University, School of Medicine, Yiwu, Zhejiang 322000, China.
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
8
|
Legutko D, Kuźniewska B, Kalita K, Yasuda R, Kaczmarek L, Michaluk P. BDNF signaling requires Matrix Metalloproteinase-9 during structural synaptic plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.08.569797. [PMID: 38106209 PMCID: PMC10723398 DOI: 10.1101/2023.12.08.569797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Synaptic plasticity underlies learning and memory processes as well as contributes, in its aberrant form, to neuropsychiatric disorders. One of its major forms is structural long-term potentiation (sLTP), an activity-dependent growth of dendritic spines that harbor excitatory synapses. The process depends on the release of brain-derived neurotrophic factor (BDNF), and activation of its receptor, TrkB. Matrix metalloproteinase-9 (MMP-9), an extracellular protease is essential for many forms of neuronal plasticity engaged in physiological as well as pathological processes. Here, we utilized two-photon microscopy and two-photon glutamate uncaging to demonstrate that MMP-9 activity is essential for sLTP and is rapidly (~seconds) released from dendritic spines in response to synaptic stimulation. Moreover, we show that either chemical or genetic inhibition of MMP-9 impairs TrkB activation, as measured by fluorescence lifetime imaging microscopy of FRET sensor. Furthermore, we provide evidence for a cell-free cleavage of proBDNF into mature BDNF by MMP-9. Our findings point to the autocrine mechanism of action of MMP-9 through BDNF maturation and TrkB activation during sLTP.
Collapse
Affiliation(s)
- Diana Legutko
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, Florida 33458, USA
| | - Bożena Kuźniewska
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
- Current address: Department of Animal Physiology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Katarzyna Kalita
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, 1 Max Planck Way, Jupiter, Florida 33458, USA
| | - Leszek Kaczmarek
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
| | - Piotr Michaluk
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute, 02-093 Warsaw, Pasteura 3, Poland
| |
Collapse
|
9
|
Matusiak M, Oziębło D, Ołdak M, Rejmak E, Kaczmarek L, Skarżyński H. Longitudinal Changes in BDNF and MMP-9 Protein Plasma Levels in Children after Cochlear Implantation. Int J Mol Sci 2023; 24:ijms24043714. [PMID: 36835126 PMCID: PMC9959301 DOI: 10.3390/ijms24043714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/06/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023] Open
Abstract
Congenitally deaf children who undergo cochlear implantation before 1 year of age develop their auditory skills faster than children who are implanted later. In this longitudinal study, a cohort of 59 implanted children were divided into two subgroups according to their ages at implantation-below or above 1 year old-and the plasma levels of matrix metalloproteinase-9 (MMP-9), brain-derived neurotrophic factor (BDNF), and pro-BDNF were measured at 0, 8, and 18 months after cochlear implant activation, while auditory development was simultaneously evaluated using the LittlEARs Questionnaire (LEAQ). A control group consisted of 49 age-matched healthy children. We identified statistically higher BDNF levels at 0 months and at the 18-month follow-ups in the younger subgroup compared to the older one and lower LEAQ scores at 0 months in the younger subgroup. Between the subgroups, there were significant differences in the changes in BDNF levels from 0 to 8 months and in LEAQ scores from 0 to 18 months. The MMP-9 levels significantly decreased from 0 to 18 months and from 0 to 8 months in both subgroups and from 8 to 18 months only in the older one. For all measured protein concentrations, significant differences were identified between the older study subgroup and the age-matched control group.
Collapse
Affiliation(s)
- Monika Matusiak
- Oto-Rhino-Laryngosurgery Clinic, Institute of Physiology and Pathology of Hearing, M Mochnackiego 10, 02-042 Warsaw, Poland
- World Hearing Centre, Mokra 17, 05-830 Nadarzyn, Poland
- Correspondence: ; Tel.: +48-223560366
| | - Dominika Oziębło
- World Hearing Centre, Mokra 17, 05-830 Nadarzyn, Poland
- Department of Genetics, Institute of Physiology and Pathology of Hearing, M Mochnackiego 10, 02-042 Warsaw, Poland
| | - Monika Ołdak
- World Hearing Centre, Mokra 17, 05-830 Nadarzyn, Poland
- Department of Genetics, Institute of Physiology and Pathology of Hearing, M Mochnackiego 10, 02-042 Warsaw, Poland
| | - Emilia Rejmak
- BRAINCITY, Nencki Institute of Experimental Biology, L Pasteura 3, 02-093 Warsaw, Poland
| | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute of Experimental Biology, L Pasteura 3, 02-093 Warsaw, Poland
| | - Henryk Skarżyński
- Oto-Rhino-Laryngosurgery Clinic, Institute of Physiology and Pathology of Hearing, M Mochnackiego 10, 02-042 Warsaw, Poland
- World Hearing Centre, Mokra 17, 05-830 Nadarzyn, Poland
| |
Collapse
|
10
|
Chen C, Fan P, Zhang L, Xue K, Hu J, Huang J, Lu W, Xu J, Xu S, Qiu G, Ran J, Gan S. Bumetanide Rescues Aquaporin-4 Depolarization via Suppressing β-Dystroglycan Cleavage and Provides Neuroprotection in Rat Retinal Ischemia-Reperfusion Injury. Neuroscience 2023; 510:95-108. [PMID: 36493910 DOI: 10.1016/j.neuroscience.2022.11.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Aquaporin-4 (AQP4) regulates retinal water homeostasis and participates in retinal oedema pathophysiology. β-dystroglycan (β-DG) is responsible for AQP4 polarization and can be cleaved by matrix metalloproteinase-9 (MMP9). Retinal oedema induced by ischemia-reperfusion (I/R) injury is an early complication. Bumetanide (BU) has potential efficacy against cytotoxic oedema. Our study investigated the effects of β-DG cleavage on AQP4 and the roles of BU in a rat retinal I/R injury model. The model was induced by applying 110 mm Hg intraocular pressure to the anterior eye chamber. BU and U0126 (a selective ERK inhibitor) were intraperitoneally administered 15 and 30 min, respectively, before I/R induction. Rhodamine isothiocyanate extravasation detection, quantitative real-time PCR, transmission electron microscopy, hematoxylin-eosin staining, immunofluorescence staining, western blotting, and TUNEL staining were performed. AQP4 lost its polarization in the retinal perivascular domain as a result of β-DG cleavage. BU rescued AQP4 depolarization, suppressed AQP4 protein expression, attenuated retinal cytotoxic oedema, and downregulated β-DG and AQP4 mRNA expression. BU suppressed glial responses and mitochondria-mediated apoptotic protein expression, including that of Caspase-3 and Cyto C, raised the Bcl-2/Bax ratio, and lowered the number of apoptotic cells in the retina. Both BU and U0126 downregulated p-ERK and MMP9 expression. Thus, BU treatment suppressed β-DG cleavage, recovered AQP4 polarization partially via inhibiting ERK/MMP9 signaling pathway, and possess potential neuroprotective efficacy in the rat retinal ischemia-reperfusion injury model.
Collapse
Affiliation(s)
- Chunyan Chen
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Ping Fan
- Department of Gynecology and Obstetrics of The Fifth People's Hospital of Chongqing, PR China
| | - Lirong Zhang
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Kaige Xue
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Jiaheng Hu
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Juan Huang
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Weitian Lu
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Jin Xu
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Shiye Xu
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Guoping Qiu
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Jianhua Ran
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China
| | - Shengwei Gan
- Institute of Neuroscience, Basic Medicine College of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
11
|
The cell adhesion protein dystroglycan affects the structural remodeling of dendritic spines. Sci Rep 2022; 12:2506. [PMID: 35169214 PMCID: PMC8847666 DOI: 10.1038/s41598-022-06462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 01/28/2022] [Indexed: 11/30/2022] Open
Abstract
Dystroglycan (DG) is a cell membrane protein that binds to the extracellular matrix in various mammalian tissues. The function of DG has been well defined in embryonic development as well as in the proper migration of differentiated neuroblasts in the central nervous system (CNS). Although DG is known to be a target for matrix metalloproteinase-9 (MMP-9), cleaved in response to enhanced synaptic activity, the role of DG in the structural remodeling of dendritic spines is still unknown. Here, we report for the first time that the deletion of DG in rat hippocampal cell cultures causes pronounced changes in the density and morphology of dendritic spines. Furthermore, we noted a decrease in laminin, one of the major extracellular partners of DG. We have also observed that the lack of DG evokes alterations in the morphological complexity of astrocytes accompanied by a decrease in the level of aquaporin 4 (AQP4), a protein located within astrocyte endfeet surrounding neuronal dendrites and synapses. Regardless of all of these changes, we did not observe any effect of DG silencing on either excitatory or inhibitory synaptic transmission. Likewise, the knockdown of DG had no effect on Psd-95 protein expression. Our results indicate that DG is involved in dendritic spine remodeling that is not functionally reflected. This may suggest the existence of unknown mechanisms that maintain proper synaptic signaling despite impaired structure of dendritic spines. Presumably, astrocytes are involved in these processes.
Collapse
|
12
|
The α-dystrobrevins play a key role in maintaining the structure and function of the extracellular matrix-significance for protein elimination failure arteriopathies. Acta Neuropathol Commun 2021; 9:171. [PMID: 34674769 PMCID: PMC8532274 DOI: 10.1186/s40478-021-01274-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
The extracellular matrix (ECM) of the cerebral vasculature provides a pathway for the flow of interstitial fluid (ISF) and solutes out of the brain by intramural periarterial drainage (IPAD). Failure of IPAD leads to protein elimination failure arteriopathies such as cerebral amyloid angiopathy (CAA). The ECM consists of a complex network of glycoproteins and proteoglycans that form distinct basement membranes (BM) around different vascular cell types. Astrocyte endfeet that are localised against the walls of blood vessels are tethered to these BMs by dystrophin associated protein complex (DPC). Alpha-dystrobrevin (α-DB) is a key dystrophin associated protein within perivascular astrocyte endfeet; its deficiency leads to a reduction in other dystrophin associated proteins, loss of AQP4 and altered ECM. In human dementia cohorts there is a positive correlation between dystrobrevin gene expression and CAA. In the present study, we test the hypotheses that (a) the positive correlation between dystrobrevin gene expression and CAA is associated with elevated expression of α-DB at glial-vascular endfeet and (b) a deficiency in α-DB results in changes to the ECM and failure of IPAD. We used human post-mortem brain tissue with different severities of CAA and transgenic α-DB deficient mice. In human post-mortem tissue we observed a significant increase in vascular α-DB with CAA (CAA vrs. Old p < 0.005, CAA vrs. Young p < 0.005). In the mouse model of α-DB deficiency, there was early modifications to vascular ECM (collagen IV and BM thickening) that translated into reduced IPAD efficiency. Our findings highlight the important role of α-DB in maintaining structure and function of ECM, particularly as a pathway for the flow of ISF and solutes out of the brain by IPAD.
Collapse
|
13
|
Extracellular Metalloproteinases in the Plasticity of Excitatory and Inhibitory Synapses. Cells 2021; 10:cells10082055. [PMID: 34440823 PMCID: PMC8391609 DOI: 10.3390/cells10082055] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Long-term synaptic plasticity is shaped by the controlled reorganization of the synaptic proteome. A key component of this process is local proteolysis performed by the family of extracellular matrix metalloproteinases (MMPs). In recent years, considerable progress was achieved in identifying extracellular proteases involved in neuroplasticity phenomena and their protein substrates. Perisynaptic metalloproteinases regulate plastic changes at synapses through the processing of extracellular and membrane proteins. MMP9 was found to play a crucial role in excitatory synapses by controlling the NMDA-dependent LTP component. In addition, MMP3 regulates the L-type calcium channel-dependent form of LTP as well as the plasticity of neuronal excitability. Both MMP9 and MMP3 were implicated in memory and learning. Moreover, altered expression or mutations of different MMPs are associated with learning deficits and psychiatric disorders, including schizophrenia, addiction, or stress response. Contrary to excitatory drive, the investigation into the role of extracellular proteolysis in inhibitory synapses is only just beginning. Herein, we review the principal mechanisms of MMP involvement in the plasticity of excitatory transmission and the recently discovered role of proteolysis in inhibitory synapses. We discuss how different matrix metalloproteinases shape dynamics and turnover of synaptic adhesome and signal transduction pathways in neurons. Finally, we discuss future challenges in exploring synapse- and plasticity-specific functions of different metalloproteinases.
Collapse
|
14
|
Salamian A, Legutko D, Nowicka K, Badyra B, Kaźmierska-Grębowska P, Caban B, Kowalczyk T, Kaczmarek L, Beroun A. Inhibition of Matrix Metalloproteinase 9 Activity Promotes Synaptogenesis in the Hippocampus. Cereb Cortex 2021; 31:3804-3819. [PMID: 33739386 PMCID: PMC8258443 DOI: 10.1093/cercor/bhab050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/21/2021] [Accepted: 02/12/2021] [Indexed: 01/07/2023] Open
Abstract
Information coding in the hippocampus relies on the interplay between various neuronal ensembles. We discovered that the application of a cholinergic agonist, carbachol (Cch), which triggers oscillatory activity in the gamma range, induces the activity of matrix metalloproteinase 9 (MMP-9)—an enzyme necessary for the maintenance of synaptic plasticity. Using electrophysiological recordings in hippocampal organotypic slices, we show that Cch potentiates the frequency of miniature inhibitory and excitatory postsynaptic currents (mIPSCs and mEPSCs, respectively) in CA1 neurons and this effect is MMP-9 dependent. Interestingly, though MMP-9 inhibition prevents the potentiation of inhibitory events, it further boosts the frequency of excitatory mEPSCs. Such enhancement of the frequency of excitatory events is a result of increased synaptogenesis onto CA1 neurons. Thus, the function of MMP-9 in cholinergically induced plasticity in the hippocampus is to maintain the fine-tuned balance between the excitatory and the inhibitory synaptic transmission.
Collapse
Affiliation(s)
- Ahmad Salamian
- Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland.,Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Diana Legutko
- Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Klaudia Nowicka
- Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Bogna Badyra
- Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Paulina Kaźmierska-Grębowska
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Bartosz Caban
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Tomasz Kowalczyk
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland
| | - Anna Beroun
- Laboratory of Neurobiology, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland.,Laboratory of Neuronal Plasticity, Nencki-EMBL Center of Excellence for Neural Plasticity and Brain Disorders: BRAINCITY, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw 02-093, Poland
| |
Collapse
|
15
|
Dou Y, Tan Y, Yu T, Ma X, Zhou Y, Zhao Y, Zhao Y, Liu X. MiR-132 down-regulates high glucose-induced β-dystroglycan degradation through Matrix Metalloproteinases-9 up-regulation in primary neurons. J Cell Mol Med 2021; 25:7783-7795. [PMID: 34160889 PMCID: PMC8358889 DOI: 10.1111/jcmm.16669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 02/24/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Cognitive dysfunction is one of the complications of diabetes. Unfortunately, there is no effective methods to block its progression currently. One of the pathophysiological mechanisms is synaptic protein damage and neuronal signal disruption because of glucose metabolism disorder. Dystroglycan protein, located in the post‐synaptic membrane of neurons, links the intracellular cytoskeleton with extracellular matrix. Abnormal expression of dystroglycan protein affects neuronal biological functions and leads to cognitive impairment. However, there are no relevant studies to observe the changes of β‐dystroglycan protein in diabetes rat brain and in primary neurons under high glucose exposure. Our data demonstrated the alterations of cognitive abilities in the diabetic rats; β‐dystroglycan protein degradation occurred in hippocampal and cortical tissues in diabetic rat brain. We further explored the mechanisms underlying of this phenomenon. When neurons are exposed to high glucose environment in long‐term period, microRNA‐132 (miR‐132) would be down‐regulated in neurons. Matrix Metalloproteinases‐9 (MMP‐9) mRNA, as a target of miR‐132, could be up‐regulated; higher expression and overlay activity of MMP‐9 protein could increase β‐DG protein degradation. In this way, β‐DG degradation may affect structure and functions among the synapses, which related to cognition decline. It may provide some theoretical basis for elucidating the molecular mechanism of diabetes‐induced cognitive dysfunction.
Collapse
Affiliation(s)
- Yunxiao Dou
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Tan
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tongya Yu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoye Ma
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuchen Zhou
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yichen Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xueyuan Liu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Chioma VC, Kruyer A, Bobadilla AC, Angelis A, Ellison Z, Hodebourg R, Scofield MD, Kalivas PW. Heroin Seeking and Extinction From Seeking Activate Matrix Metalloproteinases at Synapses on Distinct Subpopulations of Accumbens Cells. Biol Psychiatry 2021; 89:947-958. [PMID: 33579535 PMCID: PMC8434769 DOI: 10.1016/j.biopsych.2020.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Seeking addictive drugs is regulated by synaptic plasticity in the nucleus accumbens core and involves distinct plasticity in D1 and D2 receptor-expressing medium spiny neurons (D1/2-MSNs). However, it is unknown how differential plasticity between the two cell types is coordinated. Synaptic plasticity and seeking behavior induced by drug-paired cues depends not only on plasticity in the canonical pre- and postsynapse, but also on cue-induced changes in astrocytes and the extracellular matrix adjacent to the synapse. Drug cue-induced signaling in the extracellular matrix is regulated by catalytic activity of matrix metalloproteinases MMP-2,9. We hypothesized that the cell type-specific synaptic plasticity is associated with parallel cell-specific activity of MMP-2 and MMP-9. METHODS Transgenic rats were trained on a heroin self-administration protocol in which a light/tone cue was paired with heroin delivery, followed by 2 weeks of drug withdrawal, and then reinstated to heroin-conditioned cues. Confocal microscopy was used to make morphological measurements in membrane reporter-transduced D1- and D2-MSNs and astrocytes, and MMP-2,9 gelatinase activity adjacent to cell surfaces was quantified using in vivo zymography. RESULTS Presenting heroin-paired cues transiently increased MMP-9 activity around D1-MSN dendritic spines and synapse-proximal astroglial processes. Conversely, extinction training induced long-lasting increases in MMP-2 activity adjacent to D2-MSN synapses. Moreover, heroin-paired cues increased tissue inhibitor of metalloproteinases TIMP-1,2, which caused transient inhibition of MMP-2 activity around D2-MSNs during cue-induced heroin seeking. CONCLUSIONS The differential regulation of heroin seeking and extinguished seeking by different MMP subtypes on distinct cell populations poses MMP-2,9 activity as an important mediator and contributor in heroin-induced cell-specific synaptic plasticity.
Collapse
Affiliation(s)
- Vivian C Chioma
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Ana-Clara Bobadilla
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; School of Pharmacy, University of Wyoming, Laramie, Wyoming
| | - Ariana Angelis
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Zachary Ellison
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Michael D Scofield
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina; Department of Anesthesia and Perioperative Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
17
|
Lee E, Lee DH. Anterior gradient 2 is involved in the post-transcriptional regulation of β-dystroglycan. Anim Cells Syst (Seoul) 2021; 25:19-27. [PMID: 33717413 PMCID: PMC7935118 DOI: 10.1080/19768354.2020.1871405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Anterior gradient 2 (AGR2) is a protein disulfide isomerase over-expressed in numerous types of cancer. Although AGR2 plays a role in ER homeostasis, its function(s) in tumorigenesis is still elusive. Here we demonstrate that AGR2 is involved in the regulation of the β-subunit of dystroglycan (β-DG), a component of the multi-protein complex linking the extracellular matrix and cytoskeletal network. In breast cancer cells, AGR2 over-expression led to the up-regulation of β-DG but not that of α-DG, while the transcript levels of these subunits were unchanged. Conversely, the reduced expression of AGR2 caused the down-regulation of β-DG. Interestingly, induced expression of AGR2 increased the degree of co-localization of AGR2 and β-DG in the cytoplasm suggesting that AGR2 facilitates the trafficking of β-DG. In addition, AGR2 over-expression caused the re-arrangement of the actin cytoskeletal network. Presumably over-expressed AGR2 up-regulates β-DG post-transcriptionally and facilitates its trafficking, which then causes re-arrangement of the cytoskeletal network, which plays a role in the adhesion and invasion of cancer cells.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Bio and Environmental Technology, Seoul Women's University, Seoul, Korea
| | - Do Hee Lee
- Department of Bio and Environmental Technology, Seoul Women's University, Seoul, Korea
| |
Collapse
|
18
|
Pekala M, Doliwa M, Kalita K. Impact of maternal immune activation on dendritic spine development. Dev Neurobiol 2021; 81:524-545. [PMID: 33382515 DOI: 10.1002/dneu.22804] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/26/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023]
Abstract
Dendritic spines are small dendritic protrusions that harbor most excitatory synapses in the brain. The proper generation and maturation of dendritic spines are crucial for the regulation of synaptic transmission and formation of neuronal circuits. Abnormalities in dendritic spine density and morphology are common pathologies in autism and schizophrenia. According to epidemiological studies, one risk factor for these neurodevelopmental disorders is maternal infection during pregnancy. This review discusses spine alterations in animal models of maternal immune activation in the context of neurodevelopmental disorders. We describe potential mechanisms that might be responsible for prenatal infection-induced changes in the dendritic spine phenotype and behavior in offspring.
Collapse
Affiliation(s)
- Martyna Pekala
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Doliwa
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
19
|
MMP-9 Signaling Pathways That Engage Rho GTPases in Brain Plasticity. Cells 2021; 10:cells10010166. [PMID: 33467671 PMCID: PMC7830260 DOI: 10.3390/cells10010166] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023] Open
Abstract
The extracellular matrix (ECM) has been identified as a critical factor affecting synaptic function. It forms a functional scaffold that provides both the structural support and the reservoir of signaling molecules necessary for communication between cellular constituents of the central nervous system (CNS). Among numerous ECM components and modifiers that play a role in the physiological and pathological synaptic plasticity, matrix metalloproteinase 9 (MMP-9) has recently emerged as a key molecule. MMP-9 may contribute to the dynamic remodeling of structural and functional plasticity by cleaving ECM components and cell adhesion molecules. Notably, MMP-9 signaling was shown to be indispensable for long-term memory formation that requires synaptic remodeling. The core regulators of the dynamic reorganization of the actin cytoskeleton and cell adhesion are the Rho family of GTPases. These proteins have been implicated in the control of a wide range of cellular processes occurring in brain physiology and pathology. Here, we discuss the contribution of Rho GTPases to MMP-9-dependent signaling pathways in the brain. We also describe how the regulation of Rho GTPases by post-translational modifications (PTMs) can influence these processes.
Collapse
|
20
|
Pijet B, Konopka A, Rejmak E, Stefaniuk M, Khomiak D, Bulska E, Pikul S, Kaczmarek L. The matrix metalloproteinase inhibitor marimastat inhibits seizures in a model of kainic acid-induced status epilepticus. Sci Rep 2020; 10:21314. [PMID: 33277582 PMCID: PMC7718901 DOI: 10.1038/s41598-020-78341-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
An intra-hippocampus injection of kainic acid serves as a model of status epilepticus and the subsequent development of temporal lobe epilepsy. Matrix metalloproteinase-9 (MMP-9) is an enzyme that controls remodeling of the extracellular milieu under physiological and pathological conditions. In response to brain insult, MMP-9 contributes to pathological synaptic plasticity that may play a role in the progression of an epileptic condition. Marimastat is a metalloproteinase inhibitor that was tested in clinical trials of cancer. The present study assessed whether marimastat can impair the development of epilepsy. The inhibitory efficacy of marimastat was initially tested in neuronal cultures in vitro. As a marker substrate, we used nectin-3. Next, we investigated the blood–brain barrier penetration of marimastat using mass spectrometry and evaluated the therapeutic potential of marimastat against seizure outcomes. We found that marimastat inhibited the cleavage of nectin-3 in hippocampal neuronal cell cultures. Marimastat penetrated the blood–brain barrier and exerted an inhibitory effect on metalloproteinase activity in the brain. Finally, marimastat decreased some seizure parameters, such as seizure score and number, but did not directly affect status epilepticus. The long-term effects of marimastat were evident up to 6 weeks after kainic acid administration, in which marimastat still inhibited seizure duration.
Collapse
Affiliation(s)
- Barbara Pijet
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland.
| | - Anna Konopka
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-093, Warsaw, Poland
| | - Emilia Rejmak
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Marzena Stefaniuk
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Danylo Khomiak
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Ewa Bulska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-093, Warsaw, Poland
| | - Stanisław Pikul
- Pikralida Sp. z o.o., Bukowska 70/b424, 60-812, Poznań, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, BRAINCITY, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| |
Collapse
|
21
|
Bouquier N, Girard B, Aparicio Arias J, Fagni L, Bertaso F, Perroy J. Gelatinase Biosensor Reports Cellular Remodeling During Epileptogenesis. Front Synaptic Neurosci 2020; 12:15. [PMID: 32372941 PMCID: PMC7186352 DOI: 10.3389/fnsyn.2020.00015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/19/2020] [Indexed: 12/26/2022] Open
Abstract
Epileptogenesis is the gradual process responsible for converting a healthy brain into an epileptic brain. This process can be triggered by a wide range of factors, including brain injury or tumors, infections, and status epilepticus. Epileptogenesis results in aberrant synaptic plasticity, neuroinflammation and seizure-induced cell death. As Matrix Metalloproteinases (MMPs) play a crucial role in cellular plasticity by remodeling the extracellular matrix (ECM), gelatinases (MMP-2 and MMP-9) were recently highlighted as key players in epileptogenesis. In this work, we engineered a biosensor to report in situ gelatinase activity in a model of epileptogenesis. This biosensor encompasses a gelatinase-sensitive activatable cell penetrating peptide (ACPP) coupled to a TAMRA fluorophore, allowing fluorescence uptake in cells displaying endogenous gelatinase activities. In a preclinical mouse model of temporal lobe epilepsy (TLE), the intrahippocampal kainate injection, ACPPs revealed a localized distribution of gelatinase activities, refining temporal cellular changes during epileptogenesis. The activity was found particularly but not only in the ipsilateral hippocampus, starting from the CA1 area and spreading to dentate gyrus from the early stages throughout chronic epilepsy, notably in neurons and microglial cells. Thus, our work shows that ACPPs are suitable molecular imaging probes for detecting the spatiotemporal pattern of gelatinase activity during epileptogenesis, suggesting their possible use as vectors to target cellular reactive changes with treatment for epileptogenesis.
Collapse
Affiliation(s)
| | - Benoit Girard
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Laurent Fagni
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Federica Bertaso
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Perroy
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
22
|
Moretti R, Caruso P. Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? Int J Mol Sci 2020; 21:E1095. [PMID: 32046035 PMCID: PMC7036993 DOI: 10.3390/ijms21031095] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
The arteriosclerosis-dependent alteration of brain perfusion is one of the major determinants in small vessel disease, since small vessels have a pivotal role in the brain's autoregulation. Nevertheless, as far as we know, endothelium distress can potentiate the flow dysregulation and lead to subcortical vascular dementia that is related to small vessel disease (SVD), also being defined as subcortical vascular dementia (sVAD), as well as microglia activation, chronic hypoxia and hypoperfusion, vessel-tone dysregulation, altered astrocytes, and pericytes functioning blood-brain barrier disruption. The molecular basis of this pathology remains controversial. The apparent consequence (or a first event, too) is the macroscopic alteration of the neurovascular coupling. Here, we examined the possible mechanisms that lead a healthy aging process towards subcortical dementia. We remarked that SVD and white matter abnormalities related to age could be accelerated and potentiated by different vascular risk factors. Vascular function changes can be heavily influenced by genetic and epigenetic factors, which are, to the best of our knowledge, mostly unknown. Metabolic demands, active neurovascular coupling, correct glymphatic process, and adequate oxidative and inflammatory responses could be bulwarks in defense of the correct aging process; their impairments lead to a potentially catastrophic and non-reversible condition.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|
23
|
Broekaart DWM, van Scheppingen J, Anink JJ, Wierts L, van het Hof B, Jansen FE, Spliet WG, van Rijen PC, Kamphuis WW, de Vries HE, Aronica E, van Vliet EA. Increased matrix metalloproteinases expression in tuberous sclerosis complex: modulation by microRNA 146a and 147b in vitro. Neuropathol Appl Neurobiol 2020; 46:142-159. [PMID: 31183875 PMCID: PMC7217197 DOI: 10.1111/nan.12572] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/05/2019] [Indexed: 01/09/2023]
Abstract
AIM Matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors (TIMPs) control proteolysis within the extracellular matrix (ECM) of the brain. Dysfunction of this enzymatic system due to brain inflammation can disrupt the blood-brain barrier (BBB) and has been implicated in the pathogenesis of epilepsy. However, this has not been extensively studied in the epileptogenic human brain. METHODS We investigated the expression and cellular localization of major MMPs (MMP2, MMP3, MMP9 and MMP14) and TIMPs (TIMP1, TIMP2, TIMP3 and TIMP4) using quantitative real-time polymerase chain reaction (RT-PCR) and immunohistochemistry in resected epileptogenic brain tissue from patients with tuberous sclerosis complex (TSC), a severe neurodevelopmental disorder characterized by intractable epilepsy and prominent neuroinflammation. Furthermore, we determined whether anti-inflammatory microRNAs, miR146a and miR147b, which can regulate gene expression at the transcriptional level, could attenuate dysregulated MMP and TIMP expression in TSC tuber-derived astroglial cultures. RESULTS We demonstrated higher mRNA and protein expression of MMPs and TIMPs in TSC tubers compared to control and perituberal brain tissue, particularly in dysmorphic neurons and giant cells, as well as in reactive astrocytes, which was associated with BBB dysfunction. More importantly, IL-1β-induced dysregulation of MMP3, TIMP2, TIMP3 and TIMP4 could be rescued by miR146a and miR147b in tuber-derived TSC cultures. CONCLUSIONS This study provides evidence of dysregulation of the MMP/TIMP proteolytic system in TSC, which is associated with BBB dysfunction. As dysregulated MMP and TIMP expression can be ameliorated in vitro by miR146a and miR147b, these miRNAs deserve further investigation as a novel therapeutic approach.
Collapse
Affiliation(s)
- D. W. M. Broekaart
- Department of (Neuro)PathologyAmsterdam NeuroscienceAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - J. van Scheppingen
- Department of (Neuro)PathologyAmsterdam NeuroscienceAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - J. J. Anink
- Department of (Neuro)PathologyAmsterdam NeuroscienceAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - L. Wierts
- Brendinn TherapeuticsAmsterdamThe Netherlands
- Department of Molecular Cell Biology and ImmunologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - B. van het Hof
- Department of Molecular Cell Biology and ImmunologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - F. E. Jansen
- Department of Pediatric NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - W. G. Spliet
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - P. C. van Rijen
- Department of NeurosurgeryRudolf Magnus Institute for NeuroscienceUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - W. W. Kamphuis
- Brendinn TherapeuticsAmsterdamThe Netherlands
- Department of Molecular Cell Biology and ImmunologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - H. E. de Vries
- Department of Molecular Cell Biology and ImmunologyAmsterdam NeuroscienceAmsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - E. Aronica
- Department of (Neuro)PathologyAmsterdam NeuroscienceAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN)HeemstedeThe Netherlands
| | - E. A. van Vliet
- Department of (Neuro)PathologyAmsterdam NeuroscienceAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Swammerdam Institute for Life SciencesCenter for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
24
|
Weekman EM, Sudduth TL, Price BR, Woolums AE, Hawthorne D, Seaks CE, Wilcock DM. Time course of neuropathological events in hyperhomocysteinemic amyloid depositing mice reveals early neuroinflammatory changes that precede amyloid changes and cerebrovascular events. J Neuroinflammation 2019; 16:284. [PMID: 31888650 PMCID: PMC6937663 DOI: 10.1186/s12974-019-1685-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/19/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Vascular contributions to cognitive impairment and dementia (VCID) are the second leading cause of dementia behind only Alzheimer's disease (AD); however, VCID is commonly found as a co-morbidity with sporadic AD. We have previously established a mouse model of VCID by inducing hyperhomocysteinemia in both wild-type and amyloid depositing mice. While we have shown the time course of neuropathological events in the wild-type mice with hyperhomocysteinemia, the effect of amyloid deposition on this time course remains unknown; therefore, in this study, we determined the time course of neuropathological changes in our mouse model of hyperhomocysteinemia-induced VCID in amyloid depositing mice. METHODS APP/PS1 mice were placed on either a diet deficient in folate and vitamins B6 and B12 and enriched in methionine to induce hyperhomocysteinemia or a control diet for 2, 6, 10, 14, or 18 weeks. Immunohistochemistry and gene expression analysis were used to determine neuroinflammatory changes. Microhemorrhages and amyloid deposition were analyzed using histology and, finally, behavior was assessed using the 2-day radial arm water maze. RESULTS Neuroinflammation, specifically a pro-inflammatory phenotype, was the first pathological change to occur. Specifically, we see a significant increase in gene expression of tumor necrosis factor alpha, interleukin 1 beta, interleukin 6, and interleukin 12a by 6 weeks. This was followed by cognitive deficits starting at 10 weeks. Finally, there is a significant increase in the number of microhemorrhages at 14 weeks on diet as well as redistribution of amyloid from the parenchyma to the vasculature. CONCLUSIONS The time course of these pathologies points to neuroinflammation as the initial, key player in homocysteine-induced VCID co-morbid with amyloid deposition and provides a possible therapeutic target and time points.
Collapse
Affiliation(s)
- Erica M Weekman
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Tiffany L Sudduth
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Brittani R Price
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Abigail E Woolums
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Danielle Hawthorne
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Charles E Seaks
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA
| | - Donna M Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40536, USA.
| |
Collapse
|
25
|
Threat Memory Reminder Under Matrix Metalloproteinase 9 Inhibitor Doxycycline Globally Reduces Subsequent Memory Plasticity. J Neurosci 2019; 39:9424-9434. [PMID: 31615840 PMCID: PMC6867817 DOI: 10.1523/jneurosci.1285-19.2019] [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: 05/29/2019] [Revised: 08/16/2019] [Accepted: 09/11/2019] [Indexed: 11/21/2022] Open
Abstract
Associative memory can be rendered malleable by a reminder. Blocking the ensuing reconsolidation process is suggested as a therapeutic target for unwanted aversive memories. Matrix metalloproteinase-9 (MMP-9) is required for structural synapse remodeling involved in memory consolidation. Inhibiting MMP-9 with doxycycline is suggested to attenuate human threat conditioning. Here, we investigated whether MMP-9 inhibition also interferes with threat memory reconsolidation. Male and female human participants (N = 78) learned the association between two visual conditioned stimuli (CS+) and a 50% chance of an unconditioned nociceptive stimulus (US), and between CS- and the absence of US. On day 7, one CS+ was reminded without reinforcement 3.5 h after ingesting either 200 mg of doxycycline or placebo. On day 14, retention of CS memory was assessed under extinction by fear-potentiated startle. Contrary to our expectations, we observed a greater CS+/CS- difference in participants who were reminded under doxycycline compared with placebo. Participants who were reminded under placebo showed extinction learning during the retention test, which was not observed in the doxycycline group. There was no difference between the reminded and the nonreminded CS+ in either group. In contrast, during relearning after the retention test, the CS+/CS- difference was more pronounced in the placebo group than in the doxycycline group. To summarize, a single dose of doxycycline before threat memory reminder appeared to have no specific impact on reconsolidation, but to globally impair extinction learning, and threat relearning, beyond drug clearance.SIGNIFICANCE STATEMENT Matrix metalloproteinase-9 inhibition appears to attenuate memory consolidation. It could also be a target for blocking reconsolidation. Here, we test this hypothesis in human threat conditioning. We find that doxycycline has no specific impact on a reminded cue, but confers a global reduction in extinction learning and threat learning beyond the clearance of the drug. This may point toward a more long-lasting impact of doxycycline treatment on memory plasticity.
Collapse
|
26
|
Zareba-Koziol M, Bartkowiak-Kaczmarek A, Figiel I, Krzystyniak A, Wojtowicz T, Bijata M, Wlodarczyk J. Stress-induced Changes in the S-palmitoylation and S-nitrosylation of Synaptic Proteins. Mol Cell Proteomics 2019; 18:1916-1938. [PMID: 31311849 PMCID: PMC6773552 DOI: 10.1074/mcp.ra119.001581] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/12/2019] [Indexed: 11/06/2022] Open
Abstract
The precise regulation of synaptic integrity is critical for neuronal network connectivity and proper brain function. Essential aspects of the activity and localization of synaptic proteins are regulated by posttranslational modifications. S-palmitoylation is a reversible covalent modification of the cysteine with palmitate. It modulates affinity of the protein for cell membranes and membranous compartments. Intracellular palmitoylation dynamics are regulated by crosstalk with other posttranslational modifications, such as S-nitrosylation. S-nitrosylation is a covalent modification of cysteine thiol by nitric oxide and can modulate protein functions. Therefore, simultaneous identification of endogenous site-specific proteomes of both cysteine modifications under certain biological conditions offers new insights into the regulation of functional pathways. Still unclear, however, are the ways in which this crosstalk is affected in brain pathology, such as stress-related disorders. Using a newly developed mass spectrometry-based approach Palmitoylation And Nitrosylation Interplay Monitoring (PANIMoni), we analyzed the endogenous S-palmitoylation and S-nitrosylation of postsynaptic density proteins at the level of specific single cysteine in a mouse model of chronic stress. Among a total of 813 S-PALM and 620 S-NO cysteine sites that were characterized on 465 and 360 proteins, respectively, we sought to identify those that were differentially affected by stress. Our data show involvement of S-palmitoylation and S-nitrosylation crosstalk in the regulation of 122 proteins including receptors, scaffolding proteins, regulatory proteins and cytoskeletal components. Our results suggest that atypical crosstalk between the S-palmitoylation and S-nitrosylation interplay of proteins involved in synaptic transmission, protein localization and regulation of synaptic plasticity might be one of the main events associated with chronic stress disorder, leading to destabilization in synaptic networks.
Collapse
Affiliation(s)
- Monika Zareba-Koziol
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland.
| | - Anna Bartkowiak-Kaczmarek
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Izabela Figiel
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Adam Krzystyniak
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Tomasz Wojtowicz
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Monika Bijata
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Science, 02-093 Warsaw, Poland.
| |
Collapse
|
27
|
Young CNJ, Chira N, Róg J, Al-Khalidi R, Benard M, Galas L, Chan P, Vaudry D, Zablocki K, Górecki DC. Sustained activation of P2X7 induces MMP-2-evoked cleavage and functional purinoceptor inhibition. J Mol Cell Biol 2019; 10:229-242. [PMID: 28992079 DOI: 10.1093/jmcb/mjx030] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/17/2017] [Indexed: 12/11/2022] Open
Abstract
P2X7 purinoceptor promotes survival or cytotoxicity depending on extracellular adenosine triphosphate (ATP) stimulus intensity controlling its ion channel or P2X7-dependent large pore (LP) functions. Mechanisms governing this operational divergence and functional idiosyncrasy are ill-understood. We have discovered a feedback loop where sustained activation of P2X7 triggers release of active matrix metalloproteinase 2 (MMP-2), which halts ion channel and LP responses via the MMP-2-dependent receptor cleavage. This mechanism operates in cells as diverse as macrophages, dystrophic myoblasts, P2X7-transfected HEK293, and human tumour cells. Given that serum-born MMP-2 activity also blocked receptor functions, P2X7 responses in vivo may decrease in organs with permeable capillaries. Therefore, this mechanism represents an important fine-tuning of P2X7 functions, reliant on both cell-autonomous and extraneous factors. Indeed, it allowed evasion from the ATP-induced cytotoxicity in macrophages and human cancer cells with high P2X7 expression levels. Finally, we demonstrate that P2X7 ablation eliminated gelatinase activity in inflamed dystrophic muscles in vivo. Thus, P2X7 antagonists could be used as an alternative to highly toxic MMP inhibitors in treatments of inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Christopher N J Young
- School of Allied Health Sciences, Faculty of Health and Life Sciences, De Montfort University, Leicester, UK
| | - Natalia Chira
- Molecular Medicine Laboratory, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Justyna Róg
- Laboratory of Cellular Metabolism, Department of Biochemistry, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Pasteur Str., Warsaw, Poland
| | - Rasha Al-Khalidi
- Molecular Medicine Laboratory, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Magalie Benard
- PRIMACEN, Cell Imaging Platform of Normandy, Inserm, IBiSA and PISSARO Proteomic Platform, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France
| | - Ludovic Galas
- PRIMACEN, Cell Imaging Platform of Normandy, Inserm, IBiSA and PISSARO Proteomic Platform, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France
| | - Philippe Chan
- PRIMACEN, Cell Imaging Platform of Normandy, Inserm, IBiSA and PISSARO Proteomic Platform, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France
| | - David Vaudry
- PRIMACEN, Cell Imaging Platform of Normandy, Inserm, IBiSA and PISSARO Proteomic Platform, Institute for Research and Innovation in Biomedicine, University of Rouen, Mont-Saint-Aignan, France
| | - Krzysztof Zablocki
- Laboratory of Cellular Metabolism, Department of Biochemistry, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Pasteur Str., Warsaw, Poland
| | - Dariusz C Górecki
- Molecular Medicine Laboratory, Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
28
|
Chopra S, Overall CM, Dufour A. Matrix metalloproteinases in the CNS: interferons get nervous. Cell Mol Life Sci 2019; 76:3083-3095. [PMID: 31165203 PMCID: PMC11105576 DOI: 10.1007/s00018-019-03171-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) have been investigated in context of chronic inflammatory diseases and demonstrated to degrade multiple components of the extracellular matrix (ECM). However, following several disappointing MMP clinical trials, recent studies have demonstrated unexpected novel functions of MMPs in viral infections and autoimmune inflammatory diseases in unanticipated locations. Thus, MMPs play additional functions in inflammation than just ECM degradation. They can regulate the activity of chemokines and cytokines of the immune response by precise proteolytic processing resulting in activation or inactivation of signaling pathways. MMPs have been demonstrated to cleave multiple substrates of the central nervous systems (CNS) and contribute to promoting and dampening diseases of the CNS. Initially, believed to be solely promoting pathologies, more than 10 MMPs to date have been shown to have protective functions. Here, we present some of the beneficial and destructive roles of MMPs in CNS pathologies and discuss strategies for the use of MMP inhibitors.
Collapse
Affiliation(s)
- Sameeksha Chopra
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Christopher M Overall
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
- Centre for Blood Research, Vancouver, BC, V6T 1Z3, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
29
|
Beroun A, Mitra S, Michaluk P, Pijet B, Stefaniuk M, Kaczmarek L. MMPs in learning and memory and neuropsychiatric disorders. Cell Mol Life Sci 2019; 76:3207-3228. [PMID: 31172215 PMCID: PMC6647627 DOI: 10.1007/s00018-019-03180-8] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases (MMPs) are a group of over twenty proteases, operating chiefly extracellularly to cleave components of the extracellular matrix, cell adhesion molecules as well as cytokines and growth factors. By virtue of their expression and activity patterns in animal models and clinical investigations, as well as functional studies with gene knockouts and enzyme inhibitors, MMPs have been demonstrated to play a paramount role in many physiological and pathological processes in the brain. In particular, they have been shown to influence learning and memory processes, as well as major neuropsychiatric disorders such as schizophrenia, various kinds of addiction, epilepsy, fragile X syndrome, and depression. A possible link connecting all those conditions is either physiological or aberrant synaptic plasticity where some MMPs, e.g., MMP-9, have been demonstrated to contribute to the structural and functional reorganization of excitatory synapses that are located on dendritic spines. Another common theme linking the aforementioned pathological conditions is neuroinflammation and MMPs have also been shown to be important mediators of immune responses.
Collapse
Affiliation(s)
- Anna Beroun
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Piotr Michaluk
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | - Barbara Pijet
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland
| | | | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
30
|
Kobylarek D, Iwanowski P, Lewandowska Z, Limphaibool N, Szafranek S, Labrzycka A, Kozubski W. Advances in the Potential Biomarkers of Epilepsy. Front Neurol 2019; 10:685. [PMID: 31312171 PMCID: PMC6614180 DOI: 10.3389/fneur.2019.00685] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is a group of chronic neurological disorders characterized by recurrent, spontaneous, and unpredictable seizures. It is one of the most common neurological disorders, affecting tens of millions of people worldwide. Comprehensive studies on epilepsy in recent decades have revealed the complexity of epileptogenesis, in which immunological processes, epigenetic modifications, and structural changes in neuronal tissues have been identified as playing a crucial role. This review discusses the recent advances in the biomarkers of epilepsy. We evaluate the possible molecular background underlying the clinical changes observed in recent studies, focusing on therapeutic investigations, and the evidence of their safety and efficacy in the human population. This article reviews the pathophysiology of epilepsy, including recent reports on the effects of oxidative stress and hypoxia, and focuses on specific biomarkers and their clinical implications, along with further perspectives in epilepsy research.
Collapse
Affiliation(s)
- Dominik Kobylarek
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | | | | |
Collapse
|
31
|
Fournier AP, Gauberti M, Quenault A, Vivien D, Macrez R, Docagne F. Reduced spinal cord parenchymal cerebrospinal fluid circulation in experimental autoimmune encephalomyelitis. J Cereb Blood Flow Metab 2019; 39:1258-1265. [PMID: 29376456 PMCID: PMC6668509 DOI: 10.1177/0271678x18754732] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
An alteration of parenchymal cerebrospinal fluid circulation (CSF) has been proposed to take part in the pathophysiology of multiple sclerosis. By using an intragate T1-weighted high-resolution MRI of the spinal cord of freely breathing mice injected with a gadolinium chelate in the cisterna magna, we show that a parenchymal CSF circulation exists in the spinal cord, in addition to that originally described in the brain. In experimental autoimmune encephalomyelitis, a model of multiple sclerosis, we show a reduction of parenchymal CSF circulation specifically in the spinal cord but not in the brain.
Collapse
Affiliation(s)
- Antoine P Fournier
- 1 Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, Caen, France
| | - Maxime Gauberti
- 1 Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, Caen, France
| | - Aurélien Quenault
- 1 Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, Caen, France
| | - Denis Vivien
- 1 Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, Caen, France.,2 Department of Clinical Research, CHU Côte de Nacre, Caen, France
| | - Richard Macrez
- 1 Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, Caen, France.,3 Emergency Medical Service (SAMU 14), Caen University Hospital, Caen, France.,4 Emergency Department, Caen University Hospital, Caen, France
| | - Fabian Docagne
- 1 Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, Caen, France
| |
Collapse
|
32
|
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
|
33
|
Zhang X, Gu Y, Li P, Jiang A, Sheng X, Jin X, Shi Y, Li G. Matrix Metalloproteases-Mediated Cleavage on β-Dystroglycan May Play a Key Role in the Blood-Brain Barrier After Intracerebral Hemorrhage in Rats. Med Sci Monit 2019; 25:794-800. [PMID: 30686819 PMCID: PMC6362757 DOI: 10.12659/msm.908500] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND It is well documented that the Blood-Brain barrier (BBB) can be damaged by matrix metalloproteases (MMPs) after intracerebral hemorrhage (ICH), but little is known about the mechanism of this effect. MATERIAL AND METHODS We established an ICH model in rats by injecting collagenase VII into the striatum. Afterwards, intraperitoneal injection of these rats with 40 mg/kg GM6001 (a MMPs inhibitor). The effects of GM6001 on ICH were investigated by neurological severity score, brain water content, Evans blue staining, hematoxylin-eosin staining, immunohistochemical staining, and Western blot assays. RESULTS We demonstrated that the neurological damage caused by ICH was relieved at 5 and 7 days following administration of GM6001. The impaired BBB induced by ICH was improved in response to GM6001 treatment at around 3 days, as evidenced by alleviated cerebral edema, decreased Evans blue extravasation, and a reduction in inflammatory cellular infiltration. Mechanism analysis revealed that ICH induced the generation of β-dystroglycan cleavage, which could be suppressed by GM6001 treatment. Furthermore, we found that recombinant MMP2 and MMP9 triggered the cleavage of β-dystroglycan in vitro, and this action could be inhibited by GM6001 administration. CONCLUSIONS Taken together, our results suggest that MMPs-mediated cleavage on β-dystroglycan may play an important role in BBB after ICH.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Yunhe Gu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Peitong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Anqi Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Xiaomeng Sheng
- Department of Neurology, Harbin Fourth Hospital, Harbin, Heilongjiang, China (mainland)
| | - Xin Jin
- Department of Neurology, Jixi People's Hospital, Jixi, Heilongjiang, China (mainland)
| | - Yue Shi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
34
|
Price BR, Wilcock DM, Weekman EM. Hyperhomocysteinemia as a Risk Factor for Vascular Contributions to Cognitive Impairment and Dementia. Front Aging Neurosci 2018; 10:350. [PMID: 30429785 PMCID: PMC6220027 DOI: 10.3389/fnagi.2018.00350] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Behind only Alzheimer's disease, vascular contributions to cognitive impairment and dementia (VCID) is the second most common cause of dementia, affecting roughly 10-40% of dementia patients. While there is no cure for VCID, several risk factors for VCID, such as diabetes, hypertension, and stroke, have been identified. Elevated plasma levels of homocysteine, termed hyperhomocysteinemia (HHcy), are a major, yet underrecognized, risk factor for VCID. B vitamin deficiency, which is the most common cause of HHcy, is common in the elderly. With B vitamin supplementation being a relatively safe and inexpensive therapeutic, the treatment of HHcy-induced VCID would seem straightforward; however, preclinical and clinical data shows it is not. Clinical trials using B vitamin supplementation have shown conflicting results about the benefits of lowering homocysteine and issues have arisen over proper study design within the trials. Studies using cell culture and animal models have proposed several mechanisms for homocysteine-induced cognitive decline, providing other targets for therapeutics. For this review, we will focus on HHcy as a risk factor for VCID, specifically, the different mechanisms proposed for homocysteine-induced cognitive decline and the clinical trials aimed at lowering plasma homocysteine.
Collapse
Affiliation(s)
| | | | - Erica M. Weekman
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
35
|
Gorlewicz A, Kaczmarek L. Pathophysiology of Trans-Synaptic Adhesion Molecules: Implications for Epilepsy. Front Cell Dev Biol 2018; 6:119. [PMID: 30298130 PMCID: PMC6160742 DOI: 10.3389/fcell.2018.00119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 08/30/2018] [Indexed: 12/31/2022] Open
Abstract
Chemical synapses are specialized interfaces between neurons in the brain that transmit and modulate information, thereby integrating cells into multiplicity of interacting neural circuits. Cell adhesion molecules (CAMs) might form trans-synaptic complexes that are crucial for the appropriate identification of synaptic partners and further for the establishment, properties, and dynamics of synapses. When affected, trans-synaptic adhesion mechanisms play a role in synaptopathies in a variety of neuropsychiatric disorders including epilepsy. This review recapitulates current understanding of trans-synaptic interactions in pathophysiology of interneuronal connections. In particular, we discuss here the possible implications of trans-synaptic adhesion dysfunction for epilepsy.
Collapse
Affiliation(s)
- Adam Gorlewicz
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
36
|
Cooper JM, Halter KA, Prosser RA. Circadian rhythm and sleep-wake systems share the dynamic extracellular synaptic milieu. Neurobiol Sleep Circadian Rhythms 2018; 5:15-36. [PMID: 31236509 PMCID: PMC6584685 DOI: 10.1016/j.nbscr.2018.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/06/2018] [Accepted: 04/10/2018] [Indexed: 01/23/2023] Open
Abstract
The mammalian circadian and sleep-wake systems are closely aligned through their coordinated regulation of daily activity patterns. Although they differ in their anatomical organization and physiological processes, they utilize overlapping regulatory mechanisms that include an assortment of proteins and molecules interacting within the extracellular space. These extracellular factors include proteases that interact with soluble proteins, membrane-attached receptors and the extracellular matrix; and cell adhesion molecules that can form complex scaffolds connecting adjacent neurons, astrocytes and their respective intracellular cytoskeletal elements. Astrocytes also participate in the dynamic regulation of both systems through modulating neuronal appositions, the extracellular space and/or through release of gliotransmitters that can further contribute to the extracellular signaling processes. Together, these extracellular elements create a system that integrates rapid neurotransmitter signaling across longer time scales and thereby adjust neuronal signaling to reflect the daily fluctuations fundamental to both systems. Here we review what is known about these extracellular processes, focusing specifically on areas of overlap between the two systems. We also highlight questions that still need to be addressed. Although we know many of the extracellular players, far more research is needed to understand the mechanisms through which they modulate the circadian and sleep-wake systems.
Collapse
Key Words
- ADAM, A disintegrin and metalloproteinase
- AMPAR, AMPA receptor
- Astrocytes
- BDNF, brain-derived neurotrophic factor
- BMAL1, Brain and muscle Arnt-like-1 protein
- Bmal1, Brain and muscle Arnt-like-1 gene
- CAM, cell adhesion molecules
- CRY, cryptochrome protein
- Cell adhesion molecules
- Circadian rhythms
- Cry, cryptochrome gene
- DD, dark-dark
- ECM, extracellular matrix
- ECS, extracellular space
- EEG, electroencephalogram
- Endo N, endoneuraminidase N
- Extracellular proteases
- GFAP, glial fibrillary acidic protein
- IL, interleukin
- Ig, immunoglobulin
- LC, locus coeruleus
- LD, light-dark
- LH, lateral hypothalamus
- LRP-1, low density lipoprotein receptor-related protein 1
- LTP, long-term potentiation
- MMP, matrix metalloproteinases
- NCAM, neural cell adhesion molecule protein
- NMDAR, NMDA receptor
- NO, nitric oxide
- NST, nucleus of the solitary tract
- Ncam, neural cell adhesion molecule gene
- Nrl, neuroligin gene
- Nrx, neurexin gene
- P2, purine type 2 receptor
- PAI-1, plasminogen activator inhibitor-1
- PER, period protein
- PPT, peduculopontine tegmental nucleus
- PSA, polysialic acid
- Per, period gene
- REMS, rapid eye movement sleep
- RSD, REM sleep disruption
- SCN, suprachiasmatic nucleus
- SWS, slow wave sleep
- Sleep-wake system
- Suprachiasmatic nucleus
- TNF, tumor necrosis factor
- TTFL, transcriptional-translational negative feedback loop
- VIP, vasoactive intestinal polypeptide
- VLPO, ventrolateral preoptic
- VP, vasopressin
- VTA, ventral tegmental area
- dNlg4, drosophila neuroligin-4 gene
- nNOS, neuronal nitric oxide synthase gene
- nNOS, neuronal nitric oxide synthase protein
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
- uPAR, uPA receptor
Collapse
|
37
|
Aguayo FI, Pacheco AA, García-Rojo GJ, Pizarro-Bauerle JA, Doberti AV, Tejos M, García-Pérez MA, Rojas PS, Fiedler JL. Matrix Metalloproteinase 9 Displays a Particular Time Response to Acute Stress: Variation in Its Levels and Activity Distribution in Rat Hippocampus. ACS Chem Neurosci 2018; 9:945-956. [PMID: 29361213 DOI: 10.1021/acschemneuro.7b00387] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
A single stress exposure facilitates memory formation through neuroplastic processes that reshape excitatory synapses in the hippocampus, probably requiring changes in extracellular matrix components. We tested the hypothesis that matrix metalloproteinase 9 (MMP-9), an enzyme that degrades components of extracellular matrix and synaptic proteins such as β-dystroglycan (β-DG43), changes their activity and distribution in rat hippocampus during the acute stress response. After 2.5 h of restraint stress, we found (i) increased MMP-9 levels and potential activity in whole hippocampal extracts, accompanied by β-DG43 cleavage, and (ii) a significant enhancement of MMP-9 immunoreactivity in dendritic fields such as stratum radiatum and the molecular layer of hippocampus. After 24 h of stress, we found that (i) MMP-9 net activity rises at somatic field, i.e., stratum pyramidale and granule cell layers, and also at synaptic field, mainly stratum radiatum and the molecular layer of hippocampus, and (ii) hippocampal synaptoneurosome fractions are enriched with MMP-9, without variation of its potential enzymatic activity, in accordance with the constant level of cleaved β-DG43. These findings indicate that stress triggers a peculiar timing response in the MMP-9 levels, net activity, and subcellular distribution in the hippocampus, suggesting its involvement in the processing of substrates during the stress response.
Collapse
Affiliation(s)
- Felipe I. Aguayo
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Aníbal A. Pacheco
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Gonzalo J. García-Rojo
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Javier A. Pizarro-Bauerle
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ana V. Doberti
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Macarena Tejos
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - María A. García-Pérez
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Paulina S. Rojas
- Escuela de Quı́mica y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Jenny L. Fiedler
- Laboratorio de Neuroplasticidad y Neurogenética, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
38
|
Gallo FT, Katche C, Morici JF, Medina JH, Weisstaub NV. Immediate Early Genes, Memory and Psychiatric Disorders: Focus on c-Fos, Egr1 and Arc. Front Behav Neurosci 2018; 12:79. [PMID: 29755331 PMCID: PMC5932360 DOI: 10.3389/fnbeh.2018.00079] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/10/2018] [Indexed: 01/08/2023] Open
Abstract
Many psychiatric disorders, despite their specific characteristics, share deficits in the cognitive domain including executive functions, emotional control and memory. However, memory deficits have been in many cases undervalued compared with other characteristics. The expression of Immediate Early Genes (IEGs) such as, c-fos, Egr1 and arc are selectively and promptly upregulated in learning and memory among neuronal subpopulations in regions associated with these processes. Changes in expression in these genes have been observed in recognition, working and fear related memories across the brain. Despite the enormous amount of data supporting changes in their expression during learning and memory and the importance of those cognitive processes in psychiatric conditions, there are very few studies analyzing the direct implication of the IEGs in mental illnesses. In this review, we discuss the role of some of the most relevant IEGs in relation with memory processes affected in psychiatric conditions.
Collapse
Affiliation(s)
- Francisco T Gallo
- Instituto de Fisiología y Biofísica Bernardo Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Cynthia Katche
- Instituto de Biología Celular y Neurociencias (IBCN) Dr. Eduardo de Robertis, Facultad de Medicina, CONICET, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Juan F Morici
- Instituto de Fisiología y Biofísica Bernardo Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencias (IBCN) Dr. Eduardo de Robertis, Facultad de Medicina, CONICET, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.,Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos (UBA), Buenos Aires, Argentina
| | - Noelia V Weisstaub
- Instituto de Fisiología y Biofísica Bernardo Houssay, Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
39
|
Bijata M, Labus J, Guseva D, Stawarski M, Butzlaff M, Dzwonek J, Schneeberg J, Böhm K, Michaluk P, Rusakov DA, Dityatev A, Wilczyński G, Wlodarczyk J, Ponimaskin E. Synaptic Remodeling Depends on Signaling between Serotonin Receptors and the Extracellular Matrix. Cell Rep 2018; 19:1767-1782. [PMID: 28564597 DOI: 10.1016/j.celrep.2017.05.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/03/2017] [Accepted: 05/04/2017] [Indexed: 02/04/2023] Open
Abstract
Rewiring of synaptic circuitry pertinent to memory formation has been associated with morphological changes in dendritic spines and with extracellular matrix (ECM) remodeling. Here, we mechanistically link these processes by uncovering a signaling pathway involving the serotonin 5-HT7 receptor (5-HT7R), matrix metalloproteinase 9 (MMP-9), the hyaluronan receptor CD44, and the small GTPase Cdc42. We highlight a physical interaction between 5-HT7R and CD44 (identified as an MMP-9 substrate in neurons) and find that 5-HT7R stimulation increases local MMP-9 activity, triggering dendritic spine remodeling, synaptic pruning, and impairment of long-term potentiation (LTP). The underlying molecular machinery involves 5-HT7R-mediated activation of MMP-9, which leads to CD44 cleavage followed by Cdc42 activation. One important physiological consequence of this interaction includes an increase in neuronal outgrowth and elongation of dendritic spines, which might have a positive effect on complex neuronal processes (e.g., reversal learning and neuronal regeneration).
Collapse
Affiliation(s)
- Monika Bijata
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Science, Pasteura 3, Warsaw 02-093, Poland
| | - Josephine Labus
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Daria Guseva
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Michał Stawarski
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Science, Pasteura 3, Warsaw 02-093, Poland
| | - Malte Butzlaff
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Joanna Dzwonek
- Department of Neurophysiology, Nencki Institute of Experimental Biology of the Polish Academy of Science, Pasteura 3, Warsaw 02-093, Poland
| | - Jenny Schneeberg
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany; Medical Faculty, Otto von Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Katrin Böhm
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany; Medical Faculty, Otto von Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Piotr Michaluk
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Science, Pasteura 3, Warsaw 02-093, Poland; UCL Institute of Neurology, University College of London, Queen Square, London WC1N 3BG, UK
| | - Dmitri A Rusakov
- UCL Institute of Neurology, University College of London, Queen Square, London WC1N 3BG, UK
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Leipziger Str. 44, 39120 Magdeburg, Germany; Medical Faculty, Otto von Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Grzegorz Wilczyński
- Department of Neurophysiology, Nencki Institute of Experimental Biology of the Polish Academy of Science, Pasteura 3, Warsaw 02-093, Poland
| | - Jakub Wlodarczyk
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Science, Pasteura 3, Warsaw 02-093, Poland.
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Center of Physiology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
40
|
Price BR, Norris CM, Sompol P, Wilcock DM. An emerging role of astrocytes in vascular contributions to cognitive impairment and dementia. J Neurochem 2018; 144:644-650. [PMID: 29222909 DOI: 10.1111/jnc.14273] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 10/30/2017] [Accepted: 11/23/2017] [Indexed: 01/15/2023]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is understood to be the second most common cause of dementia after Alzheimer's disease, and is also a frequent comorbidity with Alzheimer's disease. While VCID is widely acknowledged as a key contributor to dementia, the mechanistic underpinnings of VCID remain poorly understood. In this review, we address the potential role of astrocytes in the pathophysiology of VCID. The vast majority of the blood vessels in the brain are surrounded by astrocytic end-feet. Given that astrocytes make up a significant proportion of the cells in the brain, and that astrocytes are usually passively connected to one another through gap junctions, we hypothesize that astrocytes are key mediators of cognitive impairment because of cerebrovascular disease. In this review, we discuss the existing body of literature regarding the role of astrocytes at the vasculature in the brain, and the known consequences of their dysfunction, as well as our hypotheses regarding the role astrocytes play in VCID. This article is part of the Special Issue "Vascular Dementia".
Collapse
Affiliation(s)
- Brittani R Price
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Christopher M Norris
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Pradoldej Sompol
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Donna M Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
41
|
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: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
42
|
Murase S, Lantz CL, Quinlan EM. Light reintroduction after dark exposure reactivates plasticity in adults via perisynaptic activation of MMP-9. eLife 2017; 6:27345. [PMID: 28875930 PMCID: PMC5630258 DOI: 10.7554/elife.27345] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022] Open
Abstract
The sensitivity of ocular dominance to regulation by monocular deprivation is the canonical model of plasticity confined to a critical period. However, we have previously shown that visual deprivation through dark exposure (DE) reactivates critical period plasticity in adults. Previous work assumed that the elimination of visual input was sufficient to enhance plasticity in the adult mouse visual cortex. In contrast, here we show that light reintroduction (LRx) after DE is responsible for the reactivation of plasticity. LRx triggers degradation of the ECM, which is blocked by pharmacological inhibition or genetic ablation of matrix metalloproteinase-9 (MMP-9). LRx induces an increase in MMP-9 activity that is perisynaptic and enriched at thalamo-cortical synapses. The reactivation of plasticity by LRx is absent in Mmp9−/− mice, and is rescued by hyaluronidase, an enzyme that degrades core ECM components. Thus, the LRx-induced increase in MMP-9 removes constraints on structural and functional plasticity in the mature cortex.
Collapse
Affiliation(s)
- Sachiko Murase
- Neuroscience and Cognitive Sciences Program, Department of Biology, University of Maryland, Maryland, United States
| | - Crystal L Lantz
- Neuroscience and Cognitive Sciences Program, Department of Biology, University of Maryland, Maryland, United States
| | - Elizabeth M Quinlan
- Neuroscience and Cognitive Sciences Program, Department of Biology, University of Maryland, Maryland, United States
| |
Collapse
|
43
|
Neuronal Dystroglycan Is Necessary for Formation and Maintenance of Functional CCK-Positive Basket Cell Terminals on Pyramidal Cells. J Neurosci 2017; 36:10296-10313. [PMID: 27707967 DOI: 10.1523/jneurosci.1823-16.2016] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/17/2016] [Indexed: 02/08/2023] Open
Abstract
Distinct types of GABAergic interneurons target different subcellular domains of pyramidal cells, thereby shaping pyramidal cell activity patterns. Whether the presynaptic heterogeneity of GABAergic innervation is mirrored by specific postsynaptic factors is largely unexplored. Here we show that dystroglycan, a protein responsible for the majority of congenital muscular dystrophies when dysfunctional, has a function at postsynaptic sites restricted to a subset of GABAergic interneurons. Conditional deletion of Dag1, encoding dystroglycan, in pyramidal cells caused loss of CCK-positive basket cell terminals in hippocampus and neocortex. PV-positive basket cell terminals were unaffected in mutant mice, demonstrating interneuron subtype-specific function of dystroglycan. Loss of dystroglycan in pyramidal cells had little influence on clustering of other GABAergic postsynaptic proteins and of glutamatergic synaptic proteins. CCK-positive terminals were not established at P21 in the absence of dystroglycan and were markedly reduced when dystroglycan was ablated in adult mice, suggesting a role for dystroglycan in both formation and maintenance of CCK-positive terminals. The necessity of neuronal dystroglycan for functional innervation by CCK-positive basket cell axon terminals was confirmed by reduced frequency of inhibitory events in pyramidal cells of dystroglycan-deficient mice and further corroborated by the inefficiency of carbachol to increase IPSC frequency in these cells. Finally, neurexin binding seems dispensable for dystroglycan function because knock-in mice expressing binding-deficient T190M dystroglycan displayed normal CCK-positive terminals. Together, we describe a novel function of dystroglycan in interneuron subtype-specific trans-synaptic signaling, revealing correlation of presynaptic and postsynaptic molecular diversity. SIGNIFICANCE STATEMENT Dystroglycan, an extracellular and transmembrane protein of the dystrophin-glycoprotein complex, is at the center of molecular studies of muscular dystrophies. Although its synaptic distribution in cortical brain regions is long established, function of dystroglycan in the synapse remained obscure. Using mice that selectively lack neuronal dystroglycan, we provide evidence that a subset of GABAergic interneurons requires dystroglycan for formation and maintenance of axonal terminals on pyramidal cells. As such, dystroglycan is the first postsynaptic GABAergic protein for which an interneuron terminal-specific function could be shown. Our findings also offer a new perspective on the mechanisms that lead to intellectual disability in muscular dystrophies without associated brain malformations.
Collapse
|
44
|
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
|
45
|
Quattromani MJ, Pruvost M, Guerreiro C, Backlund F, Englund E, Aspberg A, Jaworski T, Hakon J, Ruscher K, Kaczmarek L, Vivien D, Wieloch T. Extracellular Matrix Modulation Is Driven by Experience-Dependent Plasticity During Stroke Recovery. Mol Neurobiol 2017; 55:2196-2213. [PMID: 28290150 PMCID: PMC5840227 DOI: 10.1007/s12035-017-0461-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/16/2017] [Indexed: 11/24/2022]
Abstract
Following stroke, complete cellular death in the ischemic brain area may ensue, with remaining brain areas undergoing tissue remodelling to various degrees. Experience-dependent brain plasticity exerted through an enriched environment (EE) promotes remodelling after central nervous system injury, such as stroke. Post-stroke tissue reorganization is modulated by growth inhibitory molecules differentially expressed within the ischemic hemisphere, like chondroitin sulfate proteoglycans found in perineuronal nets (PNNs). PNNs in the neocortex predominantly enwrap parvalbumin-containing GABAergic (PV/GABA) neurons, important in sensori-information processing. Here, we investigate how extracellular matrix (ECM) proteases and their inhibitors may participate in the regulation of PNN integrity during stroke recovery. Rats were subjected to photothrombotic stroke in the motor cortex, and functional deficits were assessed at 7 days of recovery. Sham and stroked rats were housed in either standard or EE conditions for 5 days, and infarct volumes were calculated. PNNs were visualized by immunohistochemistry and counted in the somatosensory cortex of both hemispheres. mRNA expression levels of ECM proteases and protease inhibitors were assessed by RT-qPCR and their activity analyzed by gel zymography. PNNs and protease activity were also studied in brains from stroke patients where similar results were observed. EE starting 2 days after stroke and continuing for 5 days stimulated behavioral recovery of limb-placement ability without affecting infarct size. EE promoted a decrease of PNNs around PV/GABA neurons and a concomitant modulation of the proteolytic activity and mRNA expression of ECM proteases and protease inhibitors in the somatosensory cortex. This study provides molecular targets for novel therapies that could support rehabilitation of stroke patients.
Collapse
Affiliation(s)
- Miriana Jlenia Quattromani
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden.
| | - Mathilde Pruvost
- INSERM UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, F-14074, Caen, France
| | - Carla Guerreiro
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Fredrik Backlund
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Elisabet Englund
- Division of Oncology and Pathology, Lund University Hospital, 22185, Lund, Sweden
| | - Anders Aspberg
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences, Lund University, BMC C12, 22184, Lund, Sweden
| | - Tomasz Jaworski
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Jakob Hakon
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Denis Vivien
- INSERM UMR-S U919, Serine Proteases and Pathophysiology of the Neurovascular Unit, Université Caen Basse Normandie, GIP Cyceron, F-14074, Caen, France.,Department of Clinical Research, Caen University Hospital, CHU Caen, 14000, Caen, France
| | - Tadeusz Wieloch
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, 22184, Lund, Sweden
| |
Collapse
|
46
|
Nagappan-Chettiar S, Johnson-Venkatesh EM, Umemori H. Activity-dependent proteolytic cleavage of cell adhesion molecules regulates excitatory synaptic development and function. Neurosci Res 2017; 116:60-69. [PMID: 27965136 PMCID: PMC5376514 DOI: 10.1016/j.neures.2016.12.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 01/21/2023]
Abstract
Activity-dependent remodeling of neuronal connections is critical to nervous system development and function. These processes rely on the ability of synapses to detect neuronal activity and translate it into the appropriate molecular signals. One way to convert neuronal activity into downstream signaling is the proteolytic cleavage of cell adhesion molecules (CAMs). Here we review studies demonstrating the mechanisms by which proteolytic processing of CAMs direct the structural and functional remodeling of excitatory glutamatergic synapses during development and plasticity. Specifically, we examine how extracellular proteolytic cleavage of CAMs switches on or off molecular signals to 1) permit, drive, or restrict synaptic maturation during development and 2) strengthen or weaken synapses during adult plasticity. We will also examine emerging studies linking improper activity-dependent proteolytic processing of CAMs to neurological disorders such as schizophrenia, brain tumors, and Alzheimer's disease. Together these findings suggest that the regulation of activity-dependent proteolytic cleavage of CAMs is vital to proper brain development and lifelong function.
Collapse
Affiliation(s)
- Sivapratha Nagappan-Chettiar
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Erin M Johnson-Venkatesh
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Moussa C, Hebron M, Huang X, Ahn J, Rissman RA, Aisen PS, Turner RS. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer's disease. J Neuroinflammation 2017; 14:1. [PMID: 28086917 PMCID: PMC5234138 DOI: 10.1186/s12974-016-0779-0] [Citation(s) in RCA: 459] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/13/2016] [Indexed: 12/21/2022] Open
Abstract
Background Treatment of mild-moderate Alzheimer’s disease (AD) subjects (N = 119) for 52 weeks with the SIRT1 activator resveratrol (up to 1 g by mouth twice daily) attenuates progressive declines in CSF Aβ40 levels and activities of daily living (ADL) scores. Methods For this retrospective study, we examined banked CSF and plasma samples from a subset of AD subjects with CSF Aβ42 <600 ng/ml (biomarker-confirmed AD) at baseline (N = 19 resveratrol-treated and N = 19 placebo-treated). We utilized multiplex Xmap technology to measure markers of neurodegenerative disease and metalloproteinases (MMPs) in parallel in CSF and plasma samples. Results Compared to the placebo-treated group, at 52 weeks, resveratrol markedly reduced CSF MMP9 and increased macrophage-derived chemokine (MDC), interleukin (IL)-4, and fibroblast growth factor (FGF)-2. Compared to baseline, resveratrol increased plasma MMP10 and decreased IL-12P40, IL12P70, and RANTES. In this subset analysis, resveratrol treatment attenuated declines in mini-mental status examination (MMSE) scores, change in ADL (ADCS-ADL) scores, and CSF Aβ42 levels during the 52-week trial, but did not alter tau levels. Conclusions Collectively, these data suggest that resveratrol decreases CSF MMP9, modulates neuro-inflammation, and induces adaptive immunity. SIRT1 activation may be a viable target for treatment or prevention of neurodegenerative disorders. Trial registration ClinicalTrials.gov NCT01504854
Collapse
Affiliation(s)
- Charbel Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Georgetown University Medical Center, 4000 Reservoir Road, NW, Washington DC, 20057, USA.
| | - Michaeline Hebron
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Georgetown University Medical Center, 4000 Reservoir Road, NW, Washington DC, 20057, USA
| | - Xu Huang
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Georgetown University Medical Center, 4000 Reservoir Road, NW, Washington DC, 20057, USA
| | - Jaeil Ahn
- Department of Neurology, Memory Disorders Program, Translational Neurotherapeutics Program, Georgetown University, Washington DC, USA
| | - Robert A Rissman
- Department of Biostatistics, Georgetown University Medical Center, 4000 Reservoir Road, NW, Washington DC, 20057, USA
| | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute (ATRI), University of Southern California, San Diego, CA, USA
| | - R Scott Turner
- Alzheimer's Disease Cooperative Study (ADCS), Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| |
Collapse
|
48
|
Stamenkovic V, Stamenkovic S, Jaworski T, Gawlak M, Jovanovic M, Jakovcevski I, Wilczynski GM, Kaczmarek L, Schachner M, Radenovic L, Andjus PR. The extracellular matrix glycoprotein tenascin-C and matrix metalloproteinases modify cerebellar structural plasticity by exposure to an enriched environment. Brain Struct Funct 2017; 222:393-415. [PMID: 27089885 DOI: 10.1007/s00429-016-1224-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 04/04/2016] [Indexed: 02/05/2023]
Abstract
The importance of the extracellular matrix (ECM) glycoprotein tenascin-C (TnC) and the ECM degrading enzymes, matrix metalloproteinases (MMPs) -2 and -9, in cerebellar histogenesis is well established. This study aimed to examine whether there is a functional relationship between these molecules in regulating structural plasticity of the lateral deep cerebellar nucleus. To this end, starting from postnatal day 21, TnC- or MMP-9-deficient mice were exposed to an enriched environment (EE). We show that 8 weeks of exposure to EE leads to reduced lectin-based staining of perineuronal nets (PNNs), reduction in the size of GABAergic and increase in the number and size of glutamatergic synaptic terminals in wild-type mice. Conversely, TnC-deficient mice showed reduced staining of PNNs compared to wild-type mice maintained under standard conditions, and exposure to EE did not further reduce, but even slightly increased PNN staining. EE did not affect the densities of the two types of synaptic terminals in TnC-deficient mice, while the size of inhibitory, but not excitatory synaptic terminals was increased. In the time frame of 4-8 weeks, MMP-9, but not MMP-2, was observed to influence PNN remodeling and cerebellar synaptic plasticity as revealed by measurement of MMP-9 activity and colocalization with PNNs and synaptic markers. These findings were supported by observations on MMP-9-deficient mice. The present study suggests that TnC contributes to the regulation of structural plasticity in the cerebellum and that interactions between TnC and MMP-9 are likely to be important for these processes to occur.
Collapse
Affiliation(s)
- Vera Stamenkovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Stefan Stamenkovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Tomasz Jaworski
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Maciej Gawlak
- Laboratory of Physiology and Pathophysiology, Center for Preclinical Research and Technology, The Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Milos Jovanovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Igor Jakovcevski
- Experimental Neurophysiology, University Hospital Cologne, 50931, Cologne, Germany
- Experimental Neurophysiology, German Center for Neurodegenerative Diseases, 53175, Bonn, Germany
| | - Grzegorz M Wilczynski
- Laboratory of Neuromorphology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, 02-093, Warsaw, Poland
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, People's Republic of China
| | - Lidija Radenovic
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia
| | - Pavle R Andjus
- Center for Laser Microscopy, Department of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, 11000, Belgrade, Serbia.
| |
Collapse
|
49
|
Kondratiuk I, Łęski S, Urbańska M, Biecek P, Devijver H, Lechat B, Van Leuven F, Kaczmarek L, Jaworski T. GSK-3β and MMP-9 Cooperate in the Control of Dendritic Spine Morphology. Mol Neurobiol 2017; 54:200-211. [PMID: 26738851 PMCID: PMC5219889 DOI: 10.1007/s12035-015-9625-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/08/2015] [Indexed: 11/02/2022]
Abstract
Changes in the morphology of dendritic spines are prominent during learning and in different neurological and neuropsychiatric diseases, including those in which glycogen synthase kinase-3β (GSK-3β) has been implicated. Despite much evidence of the involvement of GSK-3β in functional synaptic plasticity, it is unclear how GSK-3β controls structural synaptic plasticity (i.e., the number and shape of dendritic spines). In the present study, we used two mouse models overexpressing and lacking GSK-3β in neurons to investigate how GSK-3β affects the structural plasticity of dendritic spines. Following visualization of dendritic spines with DiI dye, we found that increasing GSK-3β activity increased the number of thin spines, whereas lacking GSK-3β increased the number of stubby spines in the dentate gyrus. Under conditions of neuronal excitation, increasing GSK-3β activity caused higher activity of extracellularly acting matrix metalloproteinase-9 (MMP-9), and MMP inhibition normalized thin spines in GSK-3β overexpressing mice. Administration of the nonspecific GSK-3β inhibitor lithium in animals with active MMP-9 and animals lacking MMP-9 revealed that GSK-3β and MMP-9 act in concert to control dendritic spine morphology. Altogether, our data demonstrate that the dysregulation of GSK-3β activity has dramatic consequences on dendritic spine morphology, implicating MMP-9 as a mediator of GSK-3β-induced synaptic alterations.
Collapse
Affiliation(s)
- Ilona Kondratiuk
- Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, 3 Pasteur, 02-093, Warsaw, Poland
| | - Szymon Łęski
- Laboratory of Neuroinformatics, The Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Małgorzata Urbańska
- Laboratory of Molecular and Cellular Neurobiology, The International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Przemysław Biecek
- Faculty of Mathematics, Informatics, and Mechanics, University of Warsaw, Warsaw, Poland
| | - Herman Devijver
- Department of Human Genetics, Experimental Genetics Group - LEGTEGG, KULeuven, Leuven, Belgium
| | - Benoit Lechat
- Department of Human Genetics, Experimental Genetics Group - LEGTEGG, KULeuven, Leuven, Belgium
| | - Fred Van Leuven
- Department of Human Genetics, Experimental Genetics Group - LEGTEGG, KULeuven, Leuven, Belgium
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, 3 Pasteur, 02-093, Warsaw, Poland.
| | - Tomasz Jaworski
- Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, 3 Pasteur, 02-093, Warsaw, Poland.
| |
Collapse
|
50
|
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: 6.5] [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
|