1
|
Saxena AK, Khrolia D, Chilkoti GT, Gondode PG, Sharma T, Thakur G, Banerjee BD. Modulation of the Extracellular Signal-Regulated Protein Kinase and Tissue Inhibitors of Matrix Metalloproteases-1 Gene in Chronic Neuropathic Pain. Indian J Palliat Care 2021; 27:251-256. [PMID: 34511792 PMCID: PMC8428873 DOI: 10.25259/ijpc_339_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 11/30/2022] Open
Abstract
Objectives: The aim of this study is to study the modulation of extracellular signal-regulated protein kinase (ERK) and tissue inhibitors of matrix metalloproteases 1 (TIMP 1) gene in patients with neuropathic pain (NP). Materials and Methods: In the present, cross-sectional, observational study, 2 ml of venous baseline sample was withdrawn from all the patients with neuropathic (NP) or non NP (NNP) soon after their diagnosis or on their first visit to the pain clinic. A real-time quantitative polymerase chain reaction experiment was conducted to measure the mRNA expression of TIMP1 and ERK genes in blood samples. The Delta Ct, Delta Ct, and fold change analysis of both the genes were conducted between patients with NP and NNP. Results: A total of 285 patients with chronic pain were assessed, out of which, 153 patients had NP and 132 had NNP. The average duration of chronic pain was 11 months for 285 patients. The mRNA expression of TIMP1 gene is significantly down regulated (2.65-fold) (P (-f. 01), and the mRNA expression level of ERK is significantly up regulated (2.03-fold) (P (-f. 01) in NP patients when compared with NNP. Conclusion: The mRNA expression of TIMP1 gene is significantly down regulated, and ERK is significantly up regulated in patients with NP. Further, multicentric trials with larger sample size are recommended to confirm this finding.
Collapse
Affiliation(s)
- Ashok Kumar Saxena
- Department of Anesthesiology and Critical Care, University College of Medical Sciences and Guru Teg Bahadur Hospital, Maharashtra, India
| | - Deepanshu Khrolia
- Department of Anesthesiology and Critical Care, University College of Medical Sciences and Guru Teg Bahadur Hospital, Maharashtra, India
| | - Geetanjali T Chilkoti
- Department of Anesthesiology and Critical Care, University College of Medical Sciences and Guru Teg Bahadur Hospital, Maharashtra, India
| | - Prakash Gyandev Gondode
- Department of Anesthesiology and Critical Care, All India Institute of Medical Sciences, Nagpur, Maharashtra, India
| | - Tusha Sharma
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| | - Gaurav Thakur
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| | - Basu Dev Banerjee
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| |
Collapse
|
2
|
Sarvari S, Moakedi F, Hone E, Simpkins JW, Ren X. Mechanisms in blood-brain barrier opening and metabolism-challenged cerebrovascular ischemia with emphasis on ischemic stroke. Metab Brain Dis 2020; 35:851-868. [PMID: 32297170 PMCID: PMC7988906 DOI: 10.1007/s11011-020-00573-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
Stroke is the leading cause of disability among adults as well as the 2nd leading cause of death globally. Ischemic stroke accounts for about 85% of strokes, and currently, tissue plasminogen activator (tPA), whose therapeutic window is limited to up to 4.5 h for the appropriate population, is the only FDA approved drug in practice and medicine. After a stroke, a cascade of pathophysiological events results in the opening of the blood-brain barrier (BBB) through which further complications, disabilities, and mortality are likely to threaten the patient's health. Strikingly, tPA administration in eligible patients might cause hemorrhagic transformation and sustained damage to BBB integrity. One must, therefore, delineate upon stroke onset which cellular and molecular factors mediate BBB permeability as well as what key roles BBB rupture plays in the pathophysiology of stroke. In this review article, given our past findings of mechanisms underlying BBB opening in stroke animal models, we elucidate cellular, subcellular, and molecular factors involved in BBB permeability after ischemic stroke. The contribution of each factor to stroke severity and outcome is further discussed. Determinant factors in BBB permeability and stroke include mitochondria, miRNAs, matrix metalloproteinases (MMPs), immune cells, cytokines, chemokines, and adhesion proteins. Once these factors are interrogated and their roles in the pathophysiology of stroke are determined, novel targets for drug discovery and development can be uncovered in addition to novel therapeutic avenues for human stroke management.
Collapse
Affiliation(s)
- Sajad Sarvari
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Faezeh Moakedi
- Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Emily Hone
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA
| | - James W Simpkins
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
- Experimental Stroke Core Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA
| | - Xuefang Ren
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA.
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, USA.
- Experimental Stroke Core Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506, USA.
| |
Collapse
|
3
|
Ghasemzadeh Rahbardar M, Razavi BM, Hosseinzadeh H. Investigating the ameliorative effect of alpha-mangostin on development and existing pain in a rat model of neuropathic pain. Phytother Res 2020; 34:3211-3225. [PMID: 32592535 DOI: 10.1002/ptr.6768] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/17/2020] [Accepted: 05/24/2020] [Indexed: 01/01/2023]
Abstract
Mangosteen fruit has been used for various disorders, including pain. The effects of alpha-mangostin, the main component of mangosteen, on the neuropathic pain caused by chronic constriction injury (CCI) were evaluated in rats. In treatment groups, alpha-mangostin (10, 50, 100 mg/kg/day, i.p.) was administered from Day 0, the day of surgery, for 14 days. The degree of heat hyperalgesia, cold, and mechanical allodynia was assessed on Days 0, 3, 5, 7, 10, and 14. The lumbar spinal cord levels of MDA, GSH, inflammatory markers (TLR-4, TNF-α, MMP2, COX2, IL-1β, iNOS, and NO), apoptotic markers (Bcl-2, Bax, and caspase-3) were measured by western blot on Days 7 and 14. Rats in the CCI group showed thermal hyperalgesia, cold, and mechanical allodynia on Days 3-14. All concentrations of alpha-mangostin alleviated CCI-induced behavioral alterations. MDA level augmented and GSH level decreased in the CCI group and alpha-mangostin (50, 100 mg/kg) reversed the alterations. An enhancement in the levels of all inflammatory markers, Bax, and caspase-3 was shown on Days 7 and 14, which was controlled by alpha-mangostin (50 mg/kg). The detected antinociceptive effects of alpha-mangostin may be mediated through antioxidant, anti-inflammatory, and antiapoptotic properties.
Collapse
Affiliation(s)
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Cheng Q, Tong F, Shen Y, He C, Wang C, Ding F. Achyranthes bidentata polypeptide k improves long-term neurological outcomes through reducing downstream microvascular thrombosis in experimental ischemic stroke. Brain Res 2018; 1706:166-176. [PMID: 30414726 DOI: 10.1016/j.brainres.2018.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/04/2018] [Accepted: 11/08/2018] [Indexed: 01/07/2023]
Abstract
Achyranthes bidentata Bl. (A. bidentata) occupies an important position in traditional Chinese medicine owing to the property of promoting the circulation of blood and removing stasis. Achyranthes bidentata polypeptide k (ABPPk) is one of the active components isolated from A. bidentata. We previously demonstrated that ABPPk has potent neuroprotective effects against neuronal apoptosis both in vitro and in vivo, but the roles and mechanisms of ABPPk on long-term functional recovery after ischemic stroke remain unknown. In the current study, we investigated the neuroprotective effects of ABPPk on filament transient middle cerebral artery occlusion (tMCAO) rats and found that ABPPk reduced the infarct volume and maintained the neuronal integrity in the ischemic penumbra. Moreover, we found that ABPPk might reduce the formation of downstream microthrombus through preventing ischemic-induced oxidative damage of brain endothelial cells and activation of tissue factor (TF), plasminogen activator inhibitor-1 (PAI-1), and NF-κB. ABPPk also inhibited polymorphonuclear leukocytes (PMNs) infiltration and matrix metalloproteinase-2/-9 (MMP-2/-9) activation in the ischemic penumbra. Morris water maze, foot fault test, and modified neurological severity score were assessed for a period of 6 weeks following tMCAO. ABPPk improved long-term recognition abilities and neurological outcomes after stroke compared with saline-treated rats. Taken together, these results suggested that ABPPk is beneficial to the improvement of long-term outcomes after transient cerebral ischemia injury and can be used as a potential neuroprotective agent.
Collapse
Affiliation(s)
- Qiong Cheng
- School of Biology and Basic Medical Science, Soochow University, Suzhou, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fang Tong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Chunjiao He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Caiping Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.
| |
Collapse
|
5
|
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-containing enzymes required for homeostasis. These enzymes are an important class of drug targets as their over expression is associated with many disease states. Most of the inhibitors reported against this class of proteins have failed in clinical trials due to lack of specificity. In order to assist in drug design endeavors for MMP targets, a computationally tractable pathway is presented, comprising, (1) docking of small molecule inhibitors against the target MMPs, (2) derivation of quantum mechanical charges on the zinc ion in the active site and the amino acids coordinating with zinc including the inhibitor molecule, (3) molecular dynamics simulations on the docked ligand-MMP complexes, and (4) evaluation of binding affinities of the ligand-MMP complexes via an accurate scoring function for zinc containing metalloprotein-ligand complexes. The above pathway was applied to study the interaction of the inhibitor Batimastat with MMPs, which resulted in a high correlation between the predicted and experimental binding free energies, suggesting the potential applicability of the pathway.
Collapse
|
6
|
Jobin PG, Butler GS, Overall CM. New intracellular activities of matrix metalloproteinases shine in the moonlight. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2043-2055. [PMID: 28526562 DOI: 10.1016/j.bbamcr.2017.05.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
Adaption of a single protein to perform multiple independent functions facilitates functional plasticity of the proteome allowing a limited number of protein-coding genes to perform a multitude of cellular processes. Multifunctionality is achievable by post-translational modifications and by modulating subcellular localization. Matrix metalloproteinases (MMPs), classically viewed as degraders of the extracellular matrix (ECM) responsible for matrix protein turnover, are more recently recognized as regulators of a range of extracellular bioactive molecules including chemokines, cytokines, and their binders. However, growing evidence has convincingly identified select MMPs in intracellular compartments with unexpected physiological and pathological roles. Intracellular MMPs have both proteolytic and non-proteolytic functions, including signal transduction and transcription factor activity thereby challenging their traditional designation as extracellular proteases. This review highlights current knowledge of subcellular location and activity of these "moonlighting" MMPs. Intracellular roles herald a new era of MMP research, rejuvenating interest in targeting these proteases in therapeutic strategies. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.
Collapse
Affiliation(s)
- Parker G Jobin
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Georgina S Butler
- Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada; Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher M Overall
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada; Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
7
|
Crowell AD, King K, Deitermann A, Miranpuri GS, Resnick DK. Implication of Hypothalamus in Alleviating Spinal Cord Injury-Induced Neuropathic Pain. Ann Neurosci 2016; 23:171-175. [PMID: 27721586 DOI: 10.1159/000449183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/16/2015] [Indexed: 12/21/2022] Open
Abstract
Neuropathic pain (NP) is common among spinal cord injury (SCI) patients, and there remain clinical difficulties in treating NP due to the lack of understanding of underlying mechanisms. Extracellular proteins, such as matrix metalloproteinase and β-catenin, have been shown to be activated in the spinal cord regions following an injury, and may play a key role in contributing to NP states. While these extracellular proteins have been used as therapeutic targets in the spinal cord, there has also been evidence of up-regulation in the hypothalamus following a SCI. We hypothesize that the hypothalamus is involved in regulating NP following a SCI, and hence should be researched further to determine if it is a viable target for future therapeutic treatments.
Collapse
Affiliation(s)
- Andrew D Crowell
- Department of Neurological Surgery, University of Wisconsin Hospital and Clinics, Madison, Wis., USA
| | - Kevin King
- Department of Neurological Surgery, University of Wisconsin Hospital and Clinics, Madison, Wis., USA
| | - Annika Deitermann
- Department of Neurological Surgery, University of Wisconsin Hospital and Clinics, Madison, Wis., USA
| | - Gurwattan S Miranpuri
- Department of Neurological Surgery, University of Wisconsin Hospital and Clinics, Madison, Wis., USA
| | - Daniel K Resnick
- Department of Neurological Surgery, University of Wisconsin Hospital and Clinics, Madison, Wis., USA
| |
Collapse
|
8
|
The multifaceted role of metalloproteinases in physiological and pathological conditions in embryonic and adult brains. Prog Neurobiol 2016; 155:36-56. [PMID: 27530222 DOI: 10.1016/j.pneurobio.2016.08.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/10/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a large family of ubiquitous extracellular endopeptidases, which play important roles in a variety of physiological and pathological conditions, from the embryonic stages throughout adult life. Their extraordinary physiological "success" is due to concomitant broad substrate specificities and strict regulation of their expression, activation and inhibition levels. In recent years, MMPs have gained increasing attention as significant effectors in various aspects of central nervous system (CNS) physiology. Most importantly, they have been recognized as main players in a variety of brain disorders having different etiologies and evolution. A common aspect of these pathologies is the development of acute or chronic neuroinflammation. MMPs play an integral part in determining the result of neuroinflammation, in some cases turning its beneficial outcome into a harmful one. This review summarizes the most relevant studies concerning the physiology of MMPs, highlighting their involvement in both the developing and mature CNS, in long-lasting and acute brain diseases and, finally, in nervous system repair. Recently, a concerted effort has been made in identifying therapeutic strategies for major brain diseases by targeting MMP activities. However, from this revision of the literature appears clear that MMPs have multifaceted functional characteristics, which modulate physiological processes in multiple ways and with multiple consequences. Therefore, when choosing MMPs as possible targets, great care must be taken to evaluate the delicate balance between their activation and inhibition and to determine at which stage of the disease and at what level they become active in order maximize chances of success.
Collapse
|
9
|
Nardai S, Dobolyi A, Skopál J, Lakatos K, Merkely B, Nagy Z. Delayed Gelatinase Inhibition Induces Reticulon 4 Receptor Expression in the Peri-Infarct Cortex. J Neuropathol Exp Neurol 2016; 75:379-85. [PMID: 26945033 DOI: 10.1093/jnen/nlw011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Matrix metalloproteinase (MMP) inhibition can potentially prevent hemorrhagic transformation following cerebral infarction; however, delayed-phase MMP activity is also necessary for functional recovery after experimental stroke. We sought to identify potential mechanisms responsible for the impaired recovery associated with subacute MMP inhibition in a transient middle cerebral artery occlusion model of focal ischemia in CD rats. Gelatinase inhibition was achieved by intracerebral injection of the Fn-439 MMP inhibitor 7 days after stroke. Treatment efficacy was determined on day 9 by in situ gelatin zymography. The peri-infarct cortex was identified by triphenyl tetrazolium chloride staining, and tissue samples were dissected for TaqMan array gene-expression study. Of 84 genes known to influence poststroke regeneration, we found upregulation of mRNA for the reticulon 4 receptor (Rtn4r), a major inhibitor of regenerative nerve growth in the adult CNS, and borderline expression changes for 3 additional genes (DCC, Jun, and Ngfr). Western blot confirmed increased Rtn4r protein in the peri-infarct cortex of treated animals, and double immunolabeling showed colocalization primarily with the S100 astrocyte marker. These data suggest that increased Rtn4 receptor expression in the perilesional cortex may contribute to the impaired regeneration associated with MMP inhibition in the subacute phase of cerebral infarction.
Collapse
Affiliation(s)
- Sándor Nardai
- From the Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Hungary (SN, JS, KL, BM, ZN); National Institute of Clinical Neurosciences, Budapest, Hungary (SN, ZN); and NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary (AD)
| | - Arpád Dobolyi
- From the Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Hungary (SN, JS, KL, BM, ZN); National Institute of Clinical Neurosciences, Budapest, Hungary (SN, ZN); and NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary (AD)
| | - Judit Skopál
- From the Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Hungary (SN, JS, KL, BM, ZN); National Institute of Clinical Neurosciences, Budapest, Hungary (SN, ZN); and NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary (AD)
| | - Kinga Lakatos
- From the Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Hungary (SN, JS, KL, BM, ZN); National Institute of Clinical Neurosciences, Budapest, Hungary (SN, ZN); and NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary (AD)
| | - Béla Merkely
- From the Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Hungary (SN, JS, KL, BM, ZN); National Institute of Clinical Neurosciences, Budapest, Hungary (SN, ZN); and NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary (AD)
| | - Zoltán Nagy
- From the Department Section of Vascular Neurology, Heart and Vascular Center, Semmelweis University, Budapest, Hungary (SN, JS, KL, BM, ZN); National Institute of Clinical Neurosciences, Budapest, Hungary (SN, ZN); and NAP Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary (AD).
| |
Collapse
|
10
|
Turner RJ, Sharp FR. Implications of MMP9 for Blood Brain Barrier Disruption and Hemorrhagic Transformation Following Ischemic Stroke. Front Cell Neurosci 2016; 10:56. [PMID: 26973468 PMCID: PMC4777722 DOI: 10.3389/fncel.2016.00056] [Citation(s) in RCA: 313] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/22/2016] [Indexed: 02/03/2023] Open
Abstract
Numerous studies have documented increases in matrix metalloproteinases (MMPs), specifically MMP-9 levels following stroke, with such perturbations associated with disruption of the blood brain barrier (BBB), increased risk of hemorrhagic complications, and worsened outcome. Despite this, controversy remains as to which cells release MMP-9 at the normal and pathological BBB, with even less clarity in the context of stroke. This may be further complicated by the influence of tissue plasminogen activator (tPA) treatment. The aim of the present review is to examine the relationship between neutrophils, MMP-9 and tPA following ischemic stroke to elucidate which cells are responsible for the increases in MMP-9 and resultant barrier changes and hemorrhage observed following stroke.
Collapse
Affiliation(s)
- Renée J Turner
- Discipline of Anatomy and Pathology, Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide Adelaide, SA, Australia
| | - Frank R Sharp
- Department of Neurology, MIND Institute, University of California at Davis Medical Center Sacramento, CA, USA
| |
Collapse
|
11
|
Zhang S, Kojic L, Tsang M, Grewal P, Liu J, Namjoshi D, Wellington CL, Tetzlaff W, Cynader MS, Jia W. Distinct roles for metalloproteinases during traumatic brain injury. Neurochem Int 2016; 96:46-55. [PMID: 26939762 DOI: 10.1016/j.neuint.2016.02.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/16/2016] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Significant protease activations have been reported after traumatic brain injury (TBI). These proteases are responsible for cleavage of transmembrane proteins in neurons, glial, and endothelial cells and this results in the release of their extracellular domains (ectodomains). METHODS Two TBI models were employed here, representing both closed head injury (CHI) and open head injury (OHI). In situ zymography, immunohistochemistry, bright field and confocal microscopy, quantification of immunopositive cells and statistical analysis were applied. RESULTS We found, using in situ zymography, that gelatinase activity of matrix metalloproteinases (MMP)-2 and MMP-9 was upregulated in cortex of both injury models. Using immunohistochemistry for several MPPs (Matrix metalloproteinases) and ADAMs (disintegrin and metalloproteinases), including MMP-2, -9, ADAM-10, -17, distinct patterns of induction were observed in the two TBI models. In closed head injury, an early increase in protein expression of MMP-2, -9 and ADAM-17 was found as early as 10 min post injury in cortex and peaked at 1 h for all 4 proteases examined. In contrast, after OHI the maximal expression was observed locally neighboring the impact site, at a later time-point, as long as 24 h after the injury for MMP-2 and MMP-9. Confocal microscopy revealed colocalization of the 4 proteases with the neuronal marker NeuN in CHI, but only MMP2 colocalized with NeuN in OHI. CONCLUSIONS The findings may lead to a trauma-induced therapeutic strategy triggered soon after a primary insult to improve survival and to reduce brain damage following TBI.
Collapse
Affiliation(s)
- Si Zhang
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Luba Kojic
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Michelle Tsang
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Parampal Grewal
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.
| | - Dhananjay Namjoshi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.
| | - Max S Cynader
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - William Jia
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Oxidative DNA Damage Mediated by Intranuclear MMP Activity Is Associated with Neuronal Apoptosis in Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6927328. [PMID: 26925194 PMCID: PMC4748094 DOI: 10.1155/2016/6927328] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/26/2015] [Accepted: 12/31/2015] [Indexed: 11/18/2022]
Abstract
Evidence of the pathological roles of matrix metalloproteinases (MMPs) in various neurological disorders has made them attractive therapeutic targets. MMPs disrupt the blood-brain barrier and cause neuronal death and neuroinflammation in acute cerebral ischemia and are critical for angiogenesis during recovery. However, some challenges have to be overcome before MMPs can be further validated as drug targets in stroke injury. Identifying in vivo substrates of MMPs should greatly improve our understanding of the mechanisms of ischemic injury and is critical for providing more precise drug targets. Recent works have uncovered nontraditional roles for MMPs in the cytosol and nucleus. These have shed light on intracellular targets and biological actions of MMPs, adding additional layers of complexity for therapeutic MMP inhibition. In this review, we discussed the recent advances made in understanding nuclear location of MMPs, their regulation of intranuclear sorting, and their intranuclear proteolytic activity and substrates. In particular, we highlighted the roles of intranuclear MMPs in oxidative DNA damage, neuronal apoptosis, and neuroinflammation at an early stage of stroke insult. These novel data point to new putative MMP-mediated intranuclear actions in stroke-induced pathological processes and may lead to novel approaches to treatment of stroke and other neurological diseases.
Collapse
|
13
|
Singh T, Adekoya OA, Jayaram B. Understanding the binding of inhibitors of matrix metalloproteinases by molecular docking, quantum mechanical calculations, molecular dynamics simulations, and a MMGBSA/MMBappl study. MOLECULAR BIOSYSTEMS 2015; 11:1041-51. [PMID: 25611160 DOI: 10.1039/c5mb00003c] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Matrix metalloproteinases (MMPs) consist of a class of proteins required for normal tissue function. Their over expression is associated with many disease states and hence the interest in MMPs as drug targets. Almost all MMP inhibitors have been reported to fail in clinical trials due to lack of specificity. Zinc in the binding site of metalloproteinases performs essential biological functions and contributes to the binding affinity of inhibitors. The multiple possibilities for coordination geometry and the consequent charge on the zinc atom indicate that parameters developed are not directly transferable across different families of zinc metalloproteinases with different zinc coordination geometries, active sites and ligand architectures which makes it difficult to evaluate metal-ligand interactions. In order to assist in drug design endeavors for MMP targets, a computationally tractable pathway is presented, comprising docking of small molecule inhibitors against the target MMPs, derivation of quantum mechanical charges on the zinc ion in the active site and the amino acids coordinating with zinc including the inhibitor molecule, molecular dynamics simulations on the docked ligand-MMP complexes and evaluation of binding affinities of the ligand-MMP complexes via an accurate scoring function for zinc containing metalloprotein-ligand complexes. The above pathway was applied to study the interaction of inhibitor Batimastat with MMPs, which resulted in a high correlation between the predicted binding free energies and experiment, suggesting the potential applicability of the pathway. We then proceeded to formulate a few design principles which identify the key protein residues for generating molecules with high affinity and specificity against each of the MMPs.
Collapse
Affiliation(s)
- Tanya Singh
- Department of Chemistry, Indian Institute of Technology, Hauz Khas, New Delhi-110016, India.
| | | | | |
Collapse
|
14
|
Jablonska A, Drela K, Wojcik-Stanaszek L, Janowski M, Zalewska T, Lukomska B. Short-Lived Human Umbilical Cord-Blood-Derived Neural Stem Cells Influence the Endogenous Secretome and Increase the Number of Endogenous Neural Progenitors in a Rat Model of Lacunar Stroke. Mol Neurobiol 2015; 53:6413-6425. [PMID: 26607630 PMCID: PMC5085993 DOI: 10.1007/s12035-015-9530-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 11/08/2015] [Indexed: 12/12/2022]
Abstract
Stroke is the leading cause of severe disability, and lacunar stroke is related to cognitive decline and hemiparesis. There is no effective treatment for the majority of patients with stroke. Thus, stem cell-based regenerative medicine has drawn a growing body of attention due to the capabilities for trophic factor expression and neurogenesis enhancement. Moreover, it was shown in an experimental autoimmune encephalomyelitis (EAE) model that even short-lived stem cells can be therapeutic, and we have previously observed that phenomenon indirectly. Here, in a rat model of lacunar stroke, we investigated the molecular mechanisms underlying the positive therapeutic effects of short-lived human umbilical cord-blood-derived neural stem cells (HUCB-NSCs) through the distinct measurement of exogenous human and endogenous rat trophic factors. We have also evaluated neurogenesis and metalloproteinase activity as cellular components of therapeutic activity. As expected, we observed an increased proliferation and migration of progenitors, as well as metalloproteinase activity up to 14 days post transplantation. These changes were most prominent at the 7-day time point when we observed 30 % increases in the number of bromodeoxyuridine (BrdU)-positive cells in HUCB-NSC transplanted animals. The expression of human trophic factors was present until 7 days post transplantation, which correlated well with the survival of the human graft. For these 7 days, the level of messenger RNA (mRNA) in the analyzed trophic factors was from 300-fold for CNTF to 10,000-fold for IGF, much higher compared to constitutive expression in HUCB-NSCs in vitro. What is interesting is that there was no increase in the expression of rat trophic factors during the human graft survival, compared to that in non-transplanted animals. However, there was a prolongation of a period of increased trophic expression until 14 days post transplantation, while, in non-transplanted animals, there was a significant drop in rat trophic expression at that time point. We conclude that the positive therapeutic effect of short-lived stem cells may be related to the net increase in the amount of trophic factors (rat + human) until graft death and to the prolonged increase in rat trophic factor expression subsequently.
Collapse
Affiliation(s)
- Anna Jablonska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Drela
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Luiza Wojcik-Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Miroslaw Janowski
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Cellular Imaging Section, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
15
|
Amantea D, Micieli G, Tassorelli C, Cuartero MI, Ballesteros I, Certo M, Moro MA, Lizasoain I, Bagetta G. Rational modulation of the innate immune system for neuroprotection in ischemic stroke. Front Neurosci 2015; 9:147. [PMID: 25972779 PMCID: PMC4413676 DOI: 10.3389/fnins.2015.00147] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 04/09/2015] [Indexed: 01/08/2023] Open
Abstract
The innate immune system plays a dualistic role in the evolution of ischemic brain damage and has also been implicated in ischemic tolerance produced by different conditioning stimuli. Early after ischemia, perivascular astrocytes release cytokines and activate metalloproteases (MMPs) that contribute to blood–brain barrier (BBB) disruption and vasogenic oedema; whereas at later stages, they provide extracellular glutamate uptake, BBB regeneration and neurotrophic factors release. Similarly, early activation of microglia contributes to ischemic brain injury via the production of inflammatory cytokines, including tumor necrosis factor (TNF) and interleukin (IL)-1, reactive oxygen and nitrogen species and proteases. Nevertheless, microglia also contributes to the resolution of inflammation, by releasing IL-10 and tumor growth factor (TGF)-β, and to the late reparative processes by phagocytic activity and growth factors production. Indeed, after ischemia, microglia/macrophages differentiate toward several phenotypes: the M1 pro-inflammatory phenotype is classically activated via toll-like receptors or interferon-γ, whereas M2 phenotypes are alternatively activated by regulatory mediators, such as ILs 4, 10, 13, or TGF-β. Thus, immune cells exert a dualistic role on the evolution of ischemic brain damage, since the classic phenotypes promote injury, whereas alternatively activated M2 macrophages or N2 neutrophils prompt tissue remodeling and repair. Moreover, a subdued activation of the immune system has been involved in ischemic tolerance, since different preconditioning stimuli act via modulation of inflammatory mediators, including toll-like receptors and cytokine signaling pathways. This further underscores that the immuno-modulatory approach for the treatment of ischemic stroke should be aimed at blocking the detrimental effects, while promoting the beneficial responses of the immune reaction.
Collapse
Affiliation(s)
- Diana Amantea
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria Rende, Italy
| | | | - Cristina Tassorelli
- C. Mondino National Neurological Institute Pavia, Italy ; Department of Brain and Behavioral Sciences, University of Pavia Pavia, Italy
| | - María I Cuartero
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Iván Ballesteros
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Michelangelo Certo
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria Rende, Italy
| | - María A Moro
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid and Instituto de Investigación Hospital 12 de Octubre Madrid, Spain
| | - Giacinto Bagetta
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria Rende, Italy ; Section of Neuropharmacology of Normal and Pathological Neuronal Plasticity, University Consortium for Adaptive Disorders and Head Pain, University of Calabria Rende, Italy
| |
Collapse
|
16
|
Yang Y, Rosenberg GA. Matrix metalloproteinases as therapeutic targets for stroke. Brain Res 2015; 1623:30-8. [PMID: 25916577 DOI: 10.1016/j.brainres.2015.04.024] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 04/11/2015] [Accepted: 04/13/2015] [Indexed: 01/14/2023]
Abstract
Matrix metalloproteinases (MMPs) are important in injury and recovery in ischemic injury. They are proteolytic enzymes that degrade all components of the extracellular matrix (ECM). They are secreted in a latent form, protecting the cell from damage, but once activated induce injury prior to rapid inactivation by four tissue inhibitors to metalloproteinases (TIMPs). Normally the constitutive enzymes, MMP-2 and membrane type MMP (MMP-14), are activated in a spatially specific manner and act close to the site of activation, while the inducible enzymes, MMP-3 and MMP-9, become active through the action of free radicals and other enzymes during neuroinflammation. Because of the complex nature of the interactions with tissues during development, injury and repair, the MMPs have multiple roles, participating in the injury process in the early stages and contributing to recovery during the later stages. This dual role complicates the planning of treatment strategies. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Gary A Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
17
|
Choi DH, Kim JH, Lee KH, Kim HY, Kim YS, Choi WS, Lee J. Role of neuronal NADPH oxidase 1 in the peri-infarct regions after stroke. PLoS One 2015; 10:e0116814. [PMID: 25617620 PMCID: PMC4305324 DOI: 10.1371/journal.pone.0116814] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 12/15/2014] [Indexed: 11/18/2022] Open
Abstract
The molecular mechanism underlying the selective vulnerability of neurons to oxidative damage caused by ischemia-reperfusion (I/R) injury remains unknown. We sought to determine the role of NADPH oxidase 1 (Nox1) in cerebral I/R-induced brain injury and survival of newborn cells in the ischemic injured region. Male Wistar rats were subjected to 90 min middle cerebral artery occlusion (MCAO) followed by reperfusion. After reperfusion, infarction size, level of superoxide and 8-hydroxy-2'-deoxyguanosine (8-oxo-2dG), and Nox1 immunoreactivity were determined. RNAi-mediated knockdown of Nox1 was used to investigate the role of Nox1 in I/R-induced oxidative damage, neuronal death, motor function recovery, and ischemic neurogenesis. After I/R, Nox1 expression and 8-oxo-2dG immunoreactivity was increased in cortical neurons of the peri-infarct regions. Both infarction size and neuronal death in I/R injury were significantly reduced by adeno-associated virus (AAV)-mediated transduction of Nox1 short hairpin RNA (shRNA). AAV-mediated Nox1 knockdown enhanced functional recovery after MCAO. The level of survival and differentiation of newborn cells in the peri-infarct regions were increased by Nox1 inhibition. Our data suggest that Nox-1 may be responsible for oxidative damage to DNA, subsequent cortical neuronal degeneration, functional recovery, and regulation of ischemic neurogenesis in the peri-infarct regions after stroke.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Department of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Ji-Hye Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Kyoung-Hee Lee
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Hahn-Young Kim
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Yoon-Seong Kim
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, United States of America
| | - Wahn Soo Choi
- Department of Immunology and Physiology, Functional Genomics Institute, College of Medicine, Konkuk University, Chungju, Korea
| | - Jongmin Lee
- Department of Rehabilitation Medicine, Konkuk University School of Medicine, Seoul, Korea
- Center for Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
- * E-mail:
| |
Collapse
|
18
|
Neuropathic pain: role of inflammation, immune response, and ion channel activity in central injury mechanisms. Ann Neurosci 2014; 19:125-32. [PMID: 25205985 PMCID: PMC4117080 DOI: 10.5214/ans.0972.7531.190309] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 06/30/2012] [Accepted: 07/27/2012] [Indexed: 01/11/2023] Open
Abstract
Neuropathic pain (NP) is a significant and disabling clinical problem with very few therapeutic treatment options available. A major priority is to identify the molecular mechanisms responsible for NP. Although many seemingly relevant pathways have been identified, more research is needed before effective clinical interventions can be produced. Initial insults to the nervous system, such as spinal cord injury (SCI), are often compounded by secondary mechanisms such as inflammation, the immune response, and the changing expression of receptors and ion channels. The consequences of these secondary effects myriad and compound those elicited by the primary injury. Chronic NP syndromes following SCI can greatly complicate the clinical treatment of the primary injury and result in high comorbidity. In this review, we will describe physiological outcomes associated with SCI along with some of the mechanisms known to contribute to chronic NP development.
Collapse
|
19
|
Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat Rev Drug Discov 2014; 13:904-27. [DOI: 10.1038/nrd4390] [Citation(s) in RCA: 524] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
20
|
Mukherjee A, Swarnakar S. Implication of matrix metalloproteinases in regulating neuronal disorder. Mol Biol Rep 2014; 42:1-11. [DOI: 10.1007/s11033-014-3752-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
21
|
Greco R, Tassorelli C, Mangione AS, Levandis G, Certo M, Nappi G, Bagetta G, Blandini F, Amantea D. Neuroprotection by the PARP inhibitor PJ34 modulates cerebral and circulating RAGE levels in rats exposed to focal brain ischemia. Eur J Pharmacol 2014; 744:91-7. [PMID: 25446913 DOI: 10.1016/j.ejphar.2014.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 12/30/2022]
Abstract
The receptor for advanced glycation end products (RAGE) has a potential role as a damage-sensing molecule; however, to date, its involvement in the pathophysiology of stroke and its modulation following neuroprotective treatment are not completely understood. We have previously demonstrated that expression of distinct RAGE isoforms, recognized by different antibodies, is differentially modulated in the brain of rats subjected to focal cerebral ischemia. Here, we focus on the full-length membrane-bound RAGE isoform, showing that its expression is significantly elevated in the striatum, whereas it is reduced in the cortex of rats subjected to transient middle cerebral artery occlusion (MCAo). Notably, the reduction of cortical levels of full-length RAGE detected 24 h after reperfusion is abolished by systemic administration of a neuroprotective dose of the poly(ADP-ribose) polymerase (PARP) inhibitor, N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide (PJ34). More interestingly, a significant reduction of plasma soluble RAGE (sRAGE) occurs 24 h after reperfusion and this effect is reverted by a neuroprotective dose of PJ34. Soluble forms of RAGE, generated either by alternative splicing or by proteolysis of the full-length form, effectively bind advanced glycation end products, thereby competing with the cell surface full-length RAGE, thus providing a 'decoy' function that may counteract the adverse effects of receptor signaling in neurons and may possibly exert cytoprotective effects. Thus, our data confirm the important role of RAGE in ischemic cerebral damage and, more interestingly, suggest the potential use of sRAGE as a blood biomarker of stroke severity and of neuroprotective treatment efficacy.
Collapse
Affiliation(s)
- Rosaria Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy.
| | - Cristina Tassorelli
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy; Department of Brain and Behavior, University of Pavia, Italy
| | - Antonina Stefania Mangione
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Giovanna Levandis
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Michelangelo Certo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Giuseppe Nappi
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| |
Collapse
|
22
|
Amantea D, Certo M, Russo R, Bagetta G, Corasaniti MT, Tassorelli C. Early reperfusion injury is associated to MMP2 and IL-1β elevation in cortical neurons of rats subjected to middle cerebral artery occlusion. Neuroscience 2014; 277:755-63. [PMID: 25108165 DOI: 10.1016/j.neuroscience.2014.07.064] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 12/27/2022]
Abstract
The pathophysiological processes implicated in ischemic brain damage are strongly affected by an inflammatory reaction characterized by activation of immune cells and release of soluble mediators, including cytokines and chemokines. The pro-inflammatory cytokine interleukin (IL)-1β has been implicated in ischemic brain injury, however, to date, the mechanisms involved in the maturation of this cytokine in the ischemic brain have not been completely elucidated. We have previously suggested that matrix metalloproteinases (MMPs) may be implicated in cytokine production under pathological conditions. Here, we demonstrate that significant elevation of IL-1β occurs in the cortex as early as 1h after the beginning of reperfusion in rats subjected to 2-h middle cerebral artery occlusion (MCAo). At this early stage, we observe increased expression of IL-1β in pericallosal astroglial cells and in cortical neurons and this latter signal colocalizes with elevated gelatinolytic activity. By gel zymography, we demonstrate that the increased gelatinolytic signal at 1-h reperfusion is mainly ascribed to MMP2. Thus, MMP2 seems to contribute to early brain elevation of IL-β after transient ischemia and this mechanism may promote damage since pharmacological inhibition of gelatinases by the selective MMP2/MMP9 inhibitor V provides neuroprotection in rats subjected to transient MCAo.
Collapse
Affiliation(s)
- D Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy.
| | - M Certo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - R Russo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - G Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Italy
| | - M T Corasaniti
- Department of Health Sciences, University Magna Graecia of Catanzaro, Italy
| | - C Tassorelli
- IRCCS National Neurological Institute C. Mondino Foundation, Pavia, Italy
| |
Collapse
|
23
|
Aksenenko MB, Ruksha TG. Features of matrix metalloproteinase-2 expression in the nuclei of tumor cells of a skin melanoma. VESTNIK DERMATOLOGII I VENEROLOGII 2014. [DOI: 10.25208/0042-4609-2014-90-3-65-71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Goal of the study. To compare expression of matrix metalloproteinase-2 (MMP-2) in the nuclei and cytoplasm of tumor cells and assess the mutual relation between the localization of MMP-2 expression and different clinical and morphologic criteria in patients with a skin melanoma. Materials and methods. Tumor samples obtained from paraffin blocks taken from patients suffering from a skin melanoma and treated at the Krasnoyarsk Territorial Oncologic Dispensary served as the object of the study. Skin tissue samples taken from patients suffering from a skin melanoma (n = 44) were obtained from the Krasnoyarsk Territorial Pathological Anatomy Bureau. Immunohistochemistry was performed based on a standard technique using primary anti-matrix metalloproteinase-2 antibodies. Results. MMP-2 expression was revealed in the nuclei of tumor cells of a skin melanoma in 43.1% of cases and in the cytoplasm in 56.9% of cases. No significant differences in MMP-2 expression in the nuclei and cytoplasm were revealed (p = 0.33). Patients with nuclear expression in tumor cells are characterized by a more favorable prognosis than patients with cytoplasmic enzyme expression in tumor complexes. No dependence of the intracellular localization of MMP-2 expression on the patient sex or age, tumor localization, Clark level of invasion, Breslow’s thickness, particular features of tumor lymphocyte infiltration, tumor growth phase, histological subtype, pigmentation, tumor ulceration and tumor stage according to AJCC (American Joint Committee on Cancer) was revealed. Conclusion. MMP-2 located in the nuclei can be related to hypoxia in the tumor tissue as well as increased peroxynitrite level, which, in its turn, can have an effect on the function and regulation of matrix metalloproteinases. Moreover, MMP-2 with an atypical localization can be related to tumor cells with different biological characteristics within the same tumor, which characterize the intratumoral heterogeneity of neoplasms. The biological and clinical role of changes in the intracellular localization of the enzyme needs further explanation.
Collapse
|
24
|
Bendix I, Serdar M, Herz J, von Haefen C, Nasser F, Rohrer B, Endesfelder S, Felderhoff-Mueser U, Spies CD, Sifringer M. Inhibition of acetylcholinesterase modulates NMDA receptor antagonist mediated alterations in the developing brain. Int J Mol Sci 2014; 15:3784-98. [PMID: 24595240 PMCID: PMC3975367 DOI: 10.3390/ijms15033784] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 01/05/2023] Open
Abstract
Exposure to N-methyl-d-aspartate (NMDA) receptor antagonists has been demonstrated to induce neurodegeneration in newborn rats. However, in clinical practice the use of NMDA receptor antagonists as anesthetics and sedatives cannot always be avoided. The present study investigated the effect of the indirect cholinergic agonist physostigmine on neurotrophin expression and the extracellular matrix during NMDA receptor antagonist induced injury to the immature rat brain. The aim was to investigate matrix metalloproteinase (MMP)-2 activity, as well as expression of tissue inhibitor of metalloproteinase (TIMP)-2 and brain-derived neurotrophic factor (BDNF) after co-administration of the non-competitive NMDA receptor antagonist MK801 (dizocilpine) and the acetylcholinesterase (AChE) inhibitor physostigmine. The AChE inhibitor physostigmine ameliorated the MK801-induced reduction of BDNF mRNA and protein levels, reduced MK801-triggered MMP-2 activity and prevented decreased TIMP-2 mRNA expression. Our results indicate that AChE inhibition may prevent newborn rats from MK801-mediated brain damage by enhancing neurotrophin-associated signaling pathways and by modulating the extracellular matrix.
Collapse
Affiliation(s)
- Ivo Bendix
- Department of Pediatrics I, Neonatology, University Hospital Essen, Essen 45122, Germany.
| | - Meray Serdar
- Department of Pediatrics I, Neonatology, University Hospital Essen, Essen 45122, Germany.
| | - Josephine Herz
- Department of Pediatrics I, Neonatology, University Hospital Essen, Essen 45122, Germany.
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Fatme Nasser
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Benjamin Rohrer
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | | | - Claudia D Spies
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| | - Marco Sifringer
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin 13353, Germany.
| |
Collapse
|
25
|
Chaturvedi M, Kaczmarek L. Mmp-9 inhibition: a therapeutic strategy in ischemic stroke. Mol Neurobiol 2014; 49:563-73. [PMID: 24026771 PMCID: PMC3918117 DOI: 10.1007/s12035-013-8538-z] [Citation(s) in RCA: 223] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 08/15/2013] [Indexed: 12/16/2022]
Abstract
Ischemic stroke is a leading cause of disability worldwide. In cerebral ischemia there is an enhanced expression of matrix metallo-proteinase-9 (MMP-9), which has been associated with various complications including excitotoxicity, neuronal damage, apoptosis, blood-brain barrier (BBB) opening leading to cerebral edema, and hemorrhagic transformation. Moreover, the tissue plasminogen activator (tPA), which is the only US-FDA approved treatment of ischemic stroke, has a brief 3 to 4 h time window and it has been proposed that detrimental effects of tPA beyond the 3 h since the onset of stroke are derived from its ability to activate MMP-9 that in turn contributes to the breakdown of BBB. Therefore, the available literature suggests that MMP-9 inhibition can be of therapeutic importance in ischemic stroke. Hence, combination therapies of MMP-9 inhibitor along with tPA can be beneficial in ischemic stroke. In this review we will discuss the current status of various strategies which have shown neuroprotection and extension of thrombolytic window by directly or indirectly inhibiting MMP-9 activity. In the introductory part of the review, we briefly provide an overview on ischemic stroke, commonly used models of ischemic stroke and a role of MMP-9 in ischemia. In next part, the literature is organized as various approaches which have proven neuroprotective effects through direct or indirect decrease in MMP-9 activity, namely, using biotherapeutics, involving MMP-9 gene inhibition using viral vectors; using endogenous inhibitor of MMP-9, repurposing of old drugs such as minocycline, new chemical entities like DP-b99, and finally other approaches like therapeutic hypothermia.
Collapse
Affiliation(s)
- Mayank Chaturvedi
- Laboratory of Neurobiology, Nencki Institute, Pasteura 3, 02-093 Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute, Pasteura 3, 02-093 Warsaw, Poland
| |
Collapse
|
26
|
Inzitari D, Giusti B, Nencini P, Gori AM, Nesi M, Palumbo V, Piccardi B, Armillis A, Pracucci G, Bono G, Bovi P, Consoli D, Guidotti M, Nucera A, Massaro F, Micieli G, Orlandi G, Perini F, Tassi R, Tola MR, Sessa M, Toni D, Abbate R. MMP9 variation after thrombolysis is associated with hemorrhagic transformation of lesion and death. Stroke 2013; 44:2901-3. [PMID: 23908067 DOI: 10.1161/strokeaha.113.002274] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Experimentally, matrix metalloproteinases (MMPs) play a detrimental role related to hemorrhagic transformation and severity of an ischemic brain lesion. Tissue-type plasminogen activator (tPA) enhances such effects. This study aimed to expand clinical evidence in this connection. METHODS We measured MMPs 1, 2, 3, 7, 8, 9, and tissue inhibitors of metalloproteinases 1, 2, 4 circulating level in blood taken before and 24 hours after tPA from 327 patients (mean age, 68.9±12.1 years; median National Institutes of Health Stroke Scale, 11) with acute ischemic stroke. Delta median values ([24 hours post tPA-pre tPA]/pre tPA) of each MMP or tissue inhibitors of metalloproteinase were analyzed across subgroups of patients undergoing symptomatic intracerebral hemorrhage, 3-month death, or 3-month modified Rankin Scale score 3 to 6. RESULTS Adjusting for major clinical determinants, only matrix metalloproteinase-9 variation proved independently associated with death (odds ratio [95% confidence interval], 1.58 [1.11-2.26]; P=0.045) or symptomatic intracerebral hemorrhage (odds ratio [95% confidence interval], 1.40 [1.02-1.92]; P=0.049). Both matrix metalloproteinase-9 and tissue inhibitors of metalloproteinase-4 changes were correlated with baseline, 24 hours, and 7 days National Institutes of Health Stroke Scale (Spearman P from <0.001 to 0.040). CONCLUSIONS Our clinical evidence corroborates the detrimental role of matrix metalloproteinase-9 during ischemic stroke treated with thrombolysis, and prompts clinical trials testing agents antagonizing its effects.
Collapse
Affiliation(s)
- Domenico Inzitari
- From the Stroke and Neurology Unit, Careggi University Hospital, Florence, Italy (D.I., P.N., M.N., V.P., B.P., G.P.); Department of Experimental and Clinical Medicine, Thrombosis Centre, University of Florence, Florence, Italy (B.G., A.M.G., A.A., R.A.); Neurology Unit, Ospedale di Circolo e Fondazione Macchi, University of Insubria, Varese, Italy (G.B.); SSO Stroke Unit, U.O. Neurologia d.O., DAI di Neuroscienze, Azienda Ospedaliera Integrata, Verona, Italy (P.B.); U.O. Neurologia, G. Jazzolino Hospital, Vibo Valentia, Italy (D.C.); Neurology Unit, Valduce General Hospital, Como, Italy (M.G.); Neurology Unit, Arcispedale Santa Maria Nuova, Reggio Emilia, Italy (A.N.); Neurology Unit, Misericordia e Dolce Hospital, Prato, Italy (F.M.); Istituto Neurologico Nazionale C. Mondino, Pavia, Italy (G.M.); Department of Neurosciences, Neurological Clinic, University of Pisa, Pisa, Italy (G.O.); UOC di Neurologia e "Stroke Unit", Ospedale San Bortolo, Vicenza, Italy (F.P.); U.O.C. Stroke Unit, Dipartimento di Scienze Neurologiche e Neurosensoriali, Azienda Ospedaliera Universitaria Senese, Siena, Italy (R.T.); UO Neurologia, DAI Neuroscienze-Riabilitazione, Azienda Ospedaliera-Universitaria S. Anna, Ferrara, Italy (M.R.T.); Department of Neurology, San Raffaele Scientific Institute, Milan, Italy (M.S.); and Emergency Department Stroke Unit and Department of Neurological Sciences, Sapienza University of Rome, Rome, Italy (D.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ludewig P, Sedlacik J, Gelderblom M, Bernreuther C, Korkusuz Y, Wagener C, Gerloff C, Fiehler J, Magnus T, Horst AK. Carcinoembryonic antigen-related cell adhesion molecule 1 inhibits MMP-9-mediated blood-brain-barrier breakdown in a mouse model for ischemic stroke. Circ Res 2013; 113:1013-22. [PMID: 23780386 DOI: 10.1161/circresaha.113.301207] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
RATIONALE Blood-brain-barrier (BBB) breakdown and cerebral edema result from postischemic inflammation and contribute to mortality and morbidity after ischemic stroke. A functional role for the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in the regulation of reperfusion injury has not yet been demonstrated. OBJECTIVE We sought to identify and characterize the relevance of CEACAM1-expressing inflammatory cells in BBB breakdown and outcome after ischemic stroke in Ceacam1(-/-) and wild-type mice. METHODS AND RESULTS Focal ischemia was induced by temporary occlusion of the middle cerebral artery with a microfilament. Using MRI and Evans blue permeability assays, we observed increased stroke volumes, BBB breakdown and edema formation, reduction of cerebral perfusion, and brain atrophy in Ceacam1(-/-) mice. This translated into poor performance in neurological scoring and high poststroke-associated mortality. Elevated neutrophil influx, hyperproduction, and release of neutrophil-related matrix metalloproteinase-9 in Ceacam1(-/-) mice were confirmed by immune fluorescence, flow cytometry, zymography, and stimulation of neutrophils. Importantly, neutralization of matrix metalloproteinase-9 activity in Ceacam1(-/-) mice was sufficient to alleviate stroke sizes and improve survival to the level of CEACAM1-competent animals. Immune histochemistry of murine and human poststroke autoptic brains congruently identified abundance of CEACAM1(+)matrix metalloproteinase-9(+) neutrophils in the ischemic hemispheres. CONCLUSIONS CEACAM1 controls matrix metalloproteinase-9 secretion by neutrophils in postischemic inflammation at the BBB after stroke. We propose CEACAM1 as an important inhibitory regulator of neutrophil-mediated tissue damage and BBB breakdown in focal cerebral ischemia.
Collapse
Affiliation(s)
- Peter Ludewig
- From the Institute of Clinical Chemistry, Department of Neurology, Department of Neuropathology, and Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Lin CH, Lee HT, Lee SD, Lee W, Cho CWC, Lin SZ, Wang HJ, Okano H, Su CY, Yu YL, Hsu CY, Shyu WC. Role of HIF-1α-activated Epac1 on HSC-mediated neuroplasticity in stroke model. Neurobiol Dis 2013; 58:76-91. [PMID: 23702312 DOI: 10.1016/j.nbd.2013.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/02/2013] [Accepted: 05/10/2013] [Indexed: 12/27/2022] Open
Abstract
Exchange protein activated by cAMP-1 (Epac1) plays an important role in cell proliferation, cell survival and neuronal signaling, and activation of Epac1 in endothelial progenitor cells increases their homing to ischemic muscles and promotes neovascularization in a model of hind limb ischemia. Moreover, upregulation of Epac1 occurs during organ development and in diseases such as myocardial hypertrophy, diabetes, and Alzheimer's disease. We report here that hypoxia upregulated Epac1 through HIF-1α induction in the CD34-immunosorted human umbilical cord blood hematopoietic stem cells (hUCB(34)). Importantly, implantation of hUCB(34) subjected to hypoxia-preconditioning (HP-hUCB(34)) improved stroke outcome, more than did implantation of untreated hUCB(34), in rodents subjected to cerebral ischemia, and this required Epac1-to-matrix metalloprotease (MMP) signaling. This improved therapeutic efficacy correlated with better engraftment and differentiation of these cells in the ischemic host brain. In addition, more than did implantation of untreated HP-hUCB(34), implantation of HP-hUCB(34) improved cerebral blood flow into the ischemic brain via induction of angiogenesis, facilitated proliferation/recruitment of endogenous neural progenitor cells in the ischemic brain, and promoted neurite outgrowth following cerebral ischemia. Consistent with our proposed role of Epac1-to-MMP signaling in hypoxia-preconditioning, the above mentioned effects of implanting HP-hUCB(34) could be abolished by pharmacological inhibition and genetic disruption/deletion of Epac1 or MMPs. We have discovered a HIF-1α-to-Epac1-to-MMP signaling pathway that is required for the improved therapeutic efficacy resulting from hypoxia preconditioning of hUCB(34) in vitro prior to their implantation into the host brain in vivo.
Collapse
Affiliation(s)
- Chen-Huan Lin
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cayabyab FS, Gowribai K, Walz W. Involvement of matrix metalloproteinases-2 and -9 in the formation of a lacuna-like cerebral cavity. J Neurosci Res 2013; 91:920-33. [PMID: 23606560 DOI: 10.1002/jnr.23223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/22/2013] [Accepted: 03/01/2013] [Indexed: 12/20/2022]
Abstract
We used a modified pial vessel disruption (PVD) protocol with adult male Wistar rats to mimic small-vessel stroke in the cerebral cortex. Within 3 weeks, this lesion develops into a single lacuna-like cavity, which is fluid-filled and encapsulated by reactive astrocytes. Minocycline treatment that commences 1 hr after lesion and continues for 6 days prevents the cavitation and causes a filling of the lesion with reactive astrocytes and no barrier. Here, we determined whether inhibition of matrix metalloproteinases-2 and -9 (MMPs) mediates this minocycline action. Confocal microscopy revealed increased punctate staining of MMPs inside the lesion sites after 2 days of PVD. Astrocytes lined the lesion border but showed sparse localization inside the lesion. In contrast, increased MMP levels inside the lesion coincided with increased ED1 or OX-42 immunostaining, suggesting that MMP elevation reflected increased secretions from microglia/macrophages. Imaging analyses also revealed that minocycline administered for 2 days before animal euthanasia, significantly decreased MMP levels within the lesion. Moreover, Western blot analysis of cortical tissue extracts showed a significant 30-40% upregulation of MMPs 2 days after lesion. Minocycline administered 2 hr before the lesion significantly inhibited both MMP-9 and MMP-2 levels by ∼40%. In contrast, minocycline administered 1 hr after the lesion only decreased MMP-9 levels by ∼30%. Because MMP inhibition with batimastat injection also prevented cavity formation at 21 days, we conclude that minocycline prevented the creation of a lacuna-like cyst in the cerebral cortex by inhibiting the MMP secretion from microglia in the affected tissue.
Collapse
Affiliation(s)
- Francisco S Cayabyab
- Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | | | | |
Collapse
|
30
|
Hoehna Y, Uckermann O, Luksch H, Stefovska V, Marzahn J, Theil M, Gorkiewicz T, Gawlak M, Wilczynski GM, Kaczmarek L, Ikonomidou C. Matrix metalloproteinase 9 regulates cell death following pilocarpine-induced seizures in the developing brain. Neurobiol Dis 2012; 48:339-47. [DOI: 10.1016/j.nbd.2012.06.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 06/24/2012] [Accepted: 06/27/2012] [Indexed: 12/28/2022] Open
|
31
|
Pamenter ME, Ryu J, Hua ST, Perkins GA, Mendiola VL, Gu XQ, Ellisman MH, Haddad GG. DIDS prevents ischemic membrane degradation in cultured hippocampal neurons by inhibiting matrix metalloproteinase release. PLoS One 2012; 7:e43995. [PMID: 22937143 PMCID: PMC3427179 DOI: 10.1371/journal.pone.0043995] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/27/2012] [Indexed: 12/04/2022] Open
Abstract
During stroke, cells in the infarct core exhibit rapid failure of their permeability barriers, which releases ions and inflammatory molecules that are deleterious to nearby tissue (the penumbra). Plasma membrane degradation is key to penumbral spread and is mediated by matrix metalloproteinases (MMPs), which are released via vesicular exocytosis into the extracellular fluid in response to stress. DIDS (4,4′-diisothiocyanatostilbene-2,2′-disulphonic acid) preserves membrane integrity in neurons challenged with an in vitro ischemic penumbral mimic (ischemic solution: IS) and we asked whether this action was mediated via inhibition of MMP activity. In cultured murine hippocampal neurons challenged with IS, intracellular proMMP-2 and -9 expression increased 4–10 fold and extracellular latent and active MMP isoform expression increased 2–22 fold. MMP-mediated extracellular gelatinolytic activity increased ∼20–50 fold, causing detachment of 32.1±4.5% of cells from the matrix and extensive plasma membrane degradation (>60% of cells took up vital dyes and >60% of plasma membranes were fragmented or blebbed). DIDS abolished cellular detachment and membrane degradation in neurons and the pathology-induced extracellular expression of latent and active MMPs. DIDS similarly inhibited extracellular MMP expression and cellular detachment induced by the pro-apoptotic agent staurosporine or the general proteinase agonist 4-aminophenylmercuric acetate (APMA). Conversely, DIDS-treatment did not impair stress-induced intracellular proMMP production, nor the intracellular cleavage of proMMP-2 to the active form, suggesting DIDS interferes with the vesicular extrusion of MMPs rather than directly inhibiting proteinase expression or activation. In support of this hypothesis, an antagonist of the V-type vesicular ATPase also inhibited extracellular MMP expression to a similar degree as DIDS. In addition, in a proteinase-independent model of vesicular exocytosis, DIDS prevented stimulus-evoked release of von Willebrand Factor from human umbilical vein endothelial cells. We conclude that DIDS inhibits MMP exocytosis and through this mechanism preserves neuronal membrane integrity during pathological stress.
Collapse
Affiliation(s)
- Matthew E Pamenter
- Division of Respiratory Medicine, Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Hill JW, Poddar R, Thompson JF, Rosenberg GA, Yang Y. Intranuclear matrix metalloproteinases promote DNA damage and apoptosis induced by oxygen-glucose deprivation in neurons. Neuroscience 2012; 220:277-90. [PMID: 22710064 DOI: 10.1016/j.neuroscience.2012.06.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 06/05/2012] [Accepted: 06/06/2012] [Indexed: 11/16/2022]
Abstract
Degradation of the extracellular matrix by elevated matrix metalloproteinase (MMP) activity following ischemia/reperfusion is implicated in blood-brain barrier disruption and neuronal death. In contrast to their characterized extracellular roles, we previously reported that elevated intranuclear MMP-2 and -9 (gelatinase) activity degrades nuclear DNA repair proteins and promotes accumulation of oxidative DNA damage in neurons in rat brain at 3-h reperfusion after ischemic stroke. Here, we report that treatment with a broad-spectrum MMP inhibitor significantly reduced neuronal apoptosis in rat ischemic hemispheres at 48-h reperfusion after a 90-min middle cerebral artery occlusion (MCAO). Since extracellular gelatinases in brain tissue are known to be neurotoxic during acute stroke, the contribution of intranuclear MMP-2 and -9 activities in neurons to neuronal apoptosis has been unclear. To confirm and extend our in vivo observations, oxygen-glucose deprivation (OGD), an in vitro model of ischemia/reperfusion, was employed. Primary cortical neurons were subjected to 2-h OGD with reoxygenation. Increased intranuclear gelatinase activity was detected immediately after reoxygenation onset and was maximal at 24h, while extracellular gelatinase levels remained unchanged. We detected elevated levels of both MMP-2 and -9 in neuronal nuclear extracts and gelatinase activity in neurons co-localized primarily with MMP-2. We found a marked decrease in PARP1, XRCC1, and OGG1, and decreased PARP1 activity. Pretreatment of neurons with selective MMP-2/9 inhibitor II significantly decreased gelatinase activity and downregulation of DNA repair enzymes, decreased accumulation of oxidative DNA damage, and promoted neuronal survival after OGD. Our results confirm the nuclear localization of gelatinases and their nuclear substrates observed in an animal stroke model, further supporting a novel role for intranuclear gelatinase activity in an intrinsic apoptotic pathway in neurons during acute stroke injury.
Collapse
Affiliation(s)
- J W Hill
- University of New Mexico Health Sciences Center, Department of Neurology, Albuquerque, NM 87131, USA
| | | | | | | | | |
Collapse
|
33
|
Modulation of RAGE isoforms expression in the brain and plasma of rats exposed to transient focal cerebral ischemia. Neurochem Res 2012; 37:1508-16. [PMID: 22528836 DOI: 10.1007/s11064-012-0778-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 01/20/2012] [Accepted: 02/27/2012] [Indexed: 12/14/2022]
Abstract
Activation of RAGE (receptor for advanced glycation endproducts) and of its subtypes may play a role in neuronal damage and neuroinflammation associated with brain ischemia, though the underlying mechanisms remain unclear. In this study, we have examined by Western blotting the expression of RAGE isoforms in the cerebral cortex and striatum of Wistar rats subjected to transient (1 or 2 h) middle cerebral artery occlusion (tMCAo). The findings show that the full-length RAGE (~50 kDa) and its isoforms in the 26-43 kDa range are significantly decreased in the ischemic cortex, but not in the striatum, after 1 and 2 h tMCAo when compared to the sham group. By contrast, in the striatum, ischemia-reperfusion injury caused a significant increase of full-length RAGE and its isoforms in the 72-100 kDa range. We also investigated the soluble form of RAGE, which was significantly decreased in the plasma of rats subjected to transient or permanent MCAo. In conclusion, the present data demonstrate that regional brain expression of RAGE is differentially affected by tMCAo in rat. These modifications are accompanied by a decrease in the plasma levels of soluble RAGE, thereby suggesting a potential role for soluble RAGE as a peripheral biomarker of focal ischemia.
Collapse
|
34
|
Schomberg D, Olson JK. Immune responses of microglia in the spinal cord: Contribution to pain states. Exp Neurol 2012; 234:262-70. [DOI: 10.1016/j.expneurol.2011.12.021] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 12/05/2011] [Accepted: 12/13/2011] [Indexed: 01/24/2023]
|
35
|
Abstract
Matrix metalloproteinases (MMPs) were originally identified as matrixin proteases that act in the extracellular matrix. Recent works have uncovered nontraditional roles for MMPs in the extracellular space as well as in the cytosol and nucleus. There is strong evidence that subspecialized and compartmentalized matrixins participate in many physiological and pathological cellular processes, in which they can act as both degradative and regulatory proteases. In this review, we discuss the transcriptional and translational control of matrixin expression, their regulation of intracellular sorting, and the structural basis of activation and inhibition. In particular, we highlight the emerging roles of various matrixin forms in diseases. The activity of matrix metalloproteinases is regulated at several levels, including enzyme activation, inhibition, complex formation and compartmentalization. Most MMPs are secreted and have their function in the extracellular environment. MMPs are also found inside cells, both in the nucleus, cytosol and organelles. The role of intracellular located MMPs is still poorly understood, although recent studies have unraveled some of their functions. The localization, activation and activity of MMPs are regulated by their interactions with other proteins, proteoglycan core proteins and / or their glycosaminoglycan chains, as well as other molecules. Complexes formed between MMPs and various molecules may also include interactions with noncatalytic sites. Such exosites are regions involved in substrate processing, localized outside the active site, and are potential binding sites of specific MMP inhibitors. Knowledge about regulation of MMP activity is essential for understanding various physiological processes and pathogenesis of diseases, as well as for the development of new MMP targeting drugs.
Collapse
Affiliation(s)
- Ferdinando Mannello
- Department of Biomolecular Sciences, Section of Clinical Biochemistry, Unit of Cell Biology, University Carlo Bo of Urbino, Via O. Ubaldini 7, 61029 Urbino (PU), Italy.
| | | |
Collapse
|
36
|
Deafferentation-induced redistribution of MMP-2, but not of MMP-9, depends on the emergence of GAP-43 positive axons in the adult rat cochlear nucleus. Neural Plast 2011; 2011:859359. [PMID: 22135757 PMCID: PMC3202138 DOI: 10.1155/2011/859359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 08/17/2011] [Indexed: 12/23/2022] Open
Abstract
The matrix metalloproteinases MMP-9 and MMP-2, major modulators of the extracellular matrix (ECM), were changed in amount and distribution in the rat anteroventral cochlear nucleus (AVCN) following its sensory deafferentation by cochlear ablation. To determine what causal relationships exist between the redistribution of MMP-9 and MMP-2 and deafferentation-induced reinnervation, kainic acid was stereotaxically injected into the ventral nucleus of the trapezoid body (VNTB) prior to cochlear ablation, killing cells that deliver the growth associated protein 43 (GAP-43) into AVCN. Deafferentation-induced changes in the pattern of MMP-9 staining remained unaffected by VNTB lesions. By contrast, changes in the distribution of MMP-2 normally evoked by sensory deafferentation were reversed if GAP-43 positive axons were prevented to grow in AVCN. In conclusion, GAP-43-containing axons emerging in AVCN after cochlear ablation seem to be causal for the maintenance of MMP-2-mediated ECM remodeling.
Collapse
|
37
|
Mattila OS, Strbian D, Saksi J, Pikkarainen TO, Rantanen V, Tatlisumak T, Lindsberg PJ. Cerebral mast cells mediate blood-brain barrier disruption in acute experimental ischemic stroke through perivascular gelatinase activation. Stroke 2011; 42:3600-5. [PMID: 21980200 DOI: 10.1161/strokeaha.111.632224] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Perivascularly positioned cerebral mast cells (MC) have been shown to participate in acute blood-brain barrier disruption and expansive brain edema following experimental transient cerebral ischemia. However, the underlying molecular mechanisms remain unknown. Because proteolytic gelatinase enzymes, matrix metalloproteinases (MMP)-2 and MMP-9, are thought to have a central role in compromising the integrity of the blood-brain barrier following ischemia, we examined whether cerebral MCs influence gelatinase activity in ischemic cerebral microvasculature. METHODS Rats underwent 60 minutes of middle cerebral artery occlusion followed by 3-hour reperfusion, and were treated with a MC-stabilizing (cromoglycate), or MC-degranulating (compound 48/80) agent, or vehicle. Genetically manipulated, MC-deficient WsRc(Ws/Ws) rats and their wild-type littermates (WT) underwent the same procedures. Cerebral edema and extravasation of Evans blue albumin were measured. Gelatinase activity was visualized by in situ zymography and was quantified with computerized high-throughput image and data analysis. RESULTS Activated MCs showed secretion of gelatinase-positive granules. Genetic MC deficiency decreased global gelatinase-active area (-69%, compared with WT; P<0.001) and the mean gelatinase activity of the ischemic microvasculature (-57% compared with WT; P=0.002). MC stabilization with cromoglycate decreased the percentage of microvessels with high gelatinase activity (-36% compared with saline; P<0.05). Compound 48/80 showed increased area of in situ zymography activity in the ischemic lesion (+55% compared with saline; P<0.001). Microvascular gelatinase activity correlated with brain swelling (r=0.84; P<0.001; and r=0.61; P=0.02). CONCLUSIONS Our data demonstrate that cerebral MCs participate in regulation of acute microvascular gelatinase activation and consequent blood-brain barrier disruption following transient cerebral ischemia.
Collapse
Affiliation(s)
- Olli S Mattila
- Research Programs Unit, Molecular Neurology, Biomedicum-Helsinki, University of Helsinki, and Department of Neurology, Helsinki University Central Hospital, Haartmaninkatu 8, 00029 Helsinki, Finland.
| | | | | | | | | | | | | |
Collapse
|
38
|
Liguz-Lecznar M, Ziemka-Nalecz M, Aleksy M, Kossut M, Skangiel-Kramska J, Nowicka D. Comparison of matrix metalloproteinase activation after focal cortical ischemia in young adult and aged mice. J Neurosci Res 2011; 90:203-12. [PMID: 21922513 DOI: 10.1002/jnr.22715] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/10/2011] [Accepted: 05/13/2011] [Indexed: 11/05/2022]
Abstract
Matrix metalloproteinase (MMP) activity is implicated in the degradation of the extracellular matrix during cerebral ischemia. Although many studies have demonstrated spatiotemporal patterns of activation of gelatinases (MMP-9 and MMP-2) after ischemic stroke in young adult rodents, no data exist on MMP activity in old brains. In this study, we investigated the gelatinolytic activity in young adult (3-month-old) and aged (1-year-old) mice subjected to photothrombotic stroke. Using in situ zymography and gel zymography, we found that the basal gelatinolytic activity in the intact cerebral cortex was similar at both investigated ages. Similarly, after photothrombosis, the increased gelatinolytic response up to 7 days poststroke was the same in young and aged brains. At both ages, early activation of gelatinolysis in the ischemic core and the perilesional area was present in neuronal nuclei as revealed by colocalization of gelatinolytic product with NeuN immunostaining and DAPI. Additionally, application of specific antibodies against MMP-9 and MMP-2 revealed the increase in MMP-9 immunoreactivity in cell nuclei as early as 4 hr poststroke. No differences between young and aged mice were observed concerning the level and localization of MMP-9 immunoreactivity. The lack of age-related differences in the degree and pattern of activation of gelatinolysis after focal stroke and the lack of correspondence between the results of in situ and gel zymography suggest that extracellular proteolysis is not directly responsible for the more severe outcome of ischemic stroke in aged subjects.
Collapse
Affiliation(s)
- Monika Liguz-Lecznar
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
39
|
Wójcik-Stanaszek L, Sypecka J, Szymczak P, Ziemka-Nalecz M, Khrestchatisky M, Rivera S, Zalewska T. The potential role of metalloproteinases in neurogenesis in the gerbil hippocampus following global forebrain ischemia. PLoS One 2011; 6:e22465. [PMID: 21799862 PMCID: PMC3143139 DOI: 10.1371/journal.pone.0022465] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 06/28/2011] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) have recently been considered to be involved in the neurogenic response of adult neural stem/progenitor cells. However, there is a lack of information showing direct association between the activation of MMPs and the development of neuronal progenitor cells involving proliferation and/or further differentiation in vulnerable (Cornus Ammoni-CA1) and resistant (dentate gyrus-DG) to ischemic injury areas of the brain hippocampus. PRINCIPAL FINDINGS We showed that dynamics of MMPs activation in the dentate gyrus correlated closely with the rate of proliferation and differentiation of progenitor cells into mature neurons. In contrast, in the damaged CA1 pyramidal cells layer, despite the fact that some proliferating cells exhibited antigen specific characteristic of newborn neuronal cells, these did not attain maturity. This coincides with the low, near control-level, activity of MMPs. The above results are supported by our in vitro study showing that MMP inhibitors interfered with both the proliferation and differentiation of the human neural stem cell line derived from umbilical cord blood (HUCB-NSCs) toward the neuronal lineage. CONCLUSION Taken together, the spatial and temporal profiles of MMPs activity suggest that these proteinases could be an important component in neurogenesis-associated processes in post-ischemic brain hippocampus.
Collapse
Affiliation(s)
- Luiza Wójcik-Stanaszek
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Patrycja Szymczak
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Michel Khrestchatisky
- Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 6184, CNRS, Aix-Marseille University, Marseille, France
| | - Santiago Rivera
- Neurobiologie des Interactions Cellulaires et Neurophysiopathologie (NICN), UMR 6184, CNRS, Aix-Marseille University, Marseille, France
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
40
|
Dejonckheere E, Vandenbroucke RE, Libert C. Matrix metalloproteinases as drug targets in ischemia/reperfusion injury. Drug Discov Today 2011; 16:762-78. [PMID: 21745586 DOI: 10.1016/j.drudis.2011.06.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/24/2011] [Accepted: 06/27/2011] [Indexed: 12/11/2022]
Abstract
Deficient blood supply (ischemia) is a common consequence of some surgical procedures and certain pathologies. Once blood circulation is re-established (reperfusion), a complex series of events results in recruitment of inflammatory cells, rearrangement of the extracellular matrix and induction of cell death, which lead to organ dysfunction. Although ischemia/reperfusion (I/R) injury is an important cause of death, there is no effective therapy targeting the molecular mechanism of disease progression. Matrix metalloproteinases (MMPs), which are important regulators of many cellular activities, have a central role in disease progression after I/R injury, as suggested by numerous studies using MMP inhibitors or MMP-deficient mice. Here, we review the involvement of MMP activity in the various processes following I/R injury and the therapeutic potential of MMP inhibition.
Collapse
|
41
|
Karetko-Sysa M, Skangiel-Kramska J, Nowicka D. Disturbance of perineuronal nets in the perilesional area after photothrombosis is not associated with neuronal death. Exp Neurol 2011; 231:113-26. [PMID: 21683696 DOI: 10.1016/j.expneurol.2011.05.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Revised: 04/26/2011] [Accepted: 05/22/2011] [Indexed: 11/28/2022]
Abstract
Perineuronal nets (PNNs) are a condensed form of extracellular matrix that covers the surface of a subset of neurons. Their presence limits neuronal plasticity and may protect neurons against harmful agents. Here we analyzed the relationship between spatiotemporal changes in PNN expression and cell death markers after focal cortical photothrombotic stroke in rats. We registered a substantial decrease in PNN density using Wisteria floribunda agglutinin staining and CAT-315 and brevican immunoreactivity; the decrease occurred not only in the lesion core but also in the perilesional and remote cortex as well as in homotopic contralateral cortical regions. Fluoro Jade C and TUNEL staining in perilesional and remote areas, however, showed a low density of dying cells. Our results suggest that the PNN reduction was not a result of cellular death and could be considered an attempt to create conditions favorable for synaptic remodeling.
Collapse
Affiliation(s)
- Magdalena Karetko-Sysa
- Department of Molecular and Cellular Neurobiology, The Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | |
Collapse
|
42
|
Cybulska-Klosowicz A, Liguz-Lecznar M, Nowicka D, Ziemka-Nalecz M, Kossut M, Skangiel-Kramska J. Matrix metalloproteinase inhibition counteracts impairment of cortical experience-dependent plasticity after photothrombotic stroke. Eur J Neurosci 2011; 33:2238-46. [PMID: 21615560 DOI: 10.1111/j.1460-9568.2011.07713.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Matrix metalloproteinases (MMPs) are fine modulators of brain plasticity and pathophysiology. The inhibition of MMPs shortly after ischaemic stroke reduces the infarct size and has beneficial effects on post-stroke behavioural recovery. Our previous studies have shown that photothrombotic cortical stroke disrupts use-dependent plasticity in the neighbouring cortex. The aim of the present study was to check whether the inhibition of MMPs after photothrombosis rescued the plastic capacity of the barrel cortex. To induce plasticity in adult mice, a unilateral deprivation of all vibrissae except row C was applied. The deprivation started immediately after stroke and lasted 7 days. This procedure, in control (non-stroke) animals, results in an enlargement of functional representation of the spared row, as shown with [(14)C]2-deoxyglucose uptake mapping. In mice with stroke induced by photothrombosis in the vicinity of the barrel cortex, vibrissae deprivation did not result in an enlargement of the cortical representation of the spared row C of vibrissae, which confirmed our previous results. However, when mice were injected with the broad-spectrum inhibitor of MMPs FN-439 (10 mg/kg, i.v.) immediately before a stroke, an enlargement of the representation of the spared row similar to the enlargement found in sham mice was observed. These results indicate the involvement of MMPs in the impairment of use-dependent plasticity in the vicinity of an ischaemic lesion.
Collapse
Affiliation(s)
- A Cybulska-Klosowicz
- Department of Molecular and Cellular Neurobiology, Nencki Institute, 3 Pasteur Street, 02-093 Warsaw, Poland
| | | | | | | | | | | |
Collapse
|
43
|
Yang Y, Hill JW, Rosenberg GA. Multiple roles of metalloproteinases in neurological disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 99:241-63. [PMID: 21238938 DOI: 10.1016/b978-0-12-385504-6.00006-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Once thought to mainly act in brain to remodel the extracellular matrix, the family of metalloproteinases is important in many normal and pathological processes in the nervous system. Matrix metalloproteinases (MMPs) and A disintegrin and metalloproteinases (ADAMs) are the two major families of metalloproteinases in the brain. MMPs are comprised of several related enzymes that act on extracellular molecules. Normally, they are important in angiogenesis and neurogenesis in development. In neuroinflammatory illnesses, they disrupt the basal lamina and tight junction proteins to open the blood-brain barrier (BBB). ADAMs are important in neuroinflammation through activation of tumor necrosis factor-α (TNF-α) and their action as secretases that modulate the action of receptors on the cell surface. Four tissue inhibitors of metalloproteinases (TIMPs) are the main inhibitors of the MMPs and ADAMs. Recently, MMPs were found to affect DNA repair processes by an unexpected intranuclear action. MMPs and ADAMs have been implicated in the pathophysiology of neurodegenerative diseases such as Alzheimer's disease and vascular cognitive impairment. Growing literature on the functions of MMPs and ADAMs in the central nervous system is opening up new and exciting areas of research that may lead to novel approaches to treatment of neurological diseases.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | | | | |
Collapse
|
44
|
Cauwe B, Opdenakker G. Intracellular substrate cleavage: a novel dimension in the biochemistry, biology and pathology of matrix metalloproteinases. Crit Rev Biochem Mol Biol 2010; 45:351-423. [DOI: 10.3109/10409238.2010.501783] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
45
|
Uckermann O, Luksch H, Stefovska V, Hoehna Y, Marzahn J, Theil M, Pesic M, Górkiewicz T, Gawlak M, Wilczynski GM, Kaczmarek L, Ikonomidou C. Matrix Metalloproteinases 2 and 9 Fail to Influence Drug-Induced Neuroapoptosis in Developing Rat Brain. Neurotox Res 2010; 19:638-48. [DOI: 10.1007/s12640-010-9211-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Revised: 07/15/2010] [Accepted: 07/16/2010] [Indexed: 12/30/2022]
|
46
|
Identification of distinct cellular pools of interleukin-1β during the evolution of the neuroinflammatory response induced by transient middle cerebral artery occlusion in the brain of rat. Brain Res 2010; 1313:259-69. [DOI: 10.1016/j.brainres.2009.12.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 12/01/2009] [Accepted: 12/04/2009] [Indexed: 11/23/2022]
|
47
|
Baba Y, Yasuda O, Takemura Y, Ishikawa Y, Ohishi M, Iwanami J, Mogi M, Doe N, Horiuchi M, Maeda N, Fukuo K, Rakugi H. Timp-3 deficiency impairs cognitive function in mice. J Transl Med 2009; 89:1340-7. [PMID: 19806081 PMCID: PMC3047444 DOI: 10.1038/labinvest.2009.101] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Extracellular matrix (ECM) degradation is performed primarily by matrix metalloproteinases (MMPs). MMPs have recently been shown to regulate synaptic activity in the hippocampus and to affect memory and learning. The tissue inhibitor of metalloproteinase (Timp) is an endogenous factor that controls MMP activity by binding to the catalytic site of MMPs. At present, four Timp isotypes have been reported (Timp-1 through Timp-4) with 35-50% amino-acid sequence homology. Timp-3 is a unique member of Timp proteins in that it is bound to the ECM. In this study, we used the passive avoidance test, active avoidance test, and water maze test to examine the cognitive function in Timp-3 knockout (KO) mice. Habituation was evaluated using the open-field test. The water maze test showed that Timp-3 KO mice exhibit deterioration in cognitive function compared with wild-type (WT) mice. The open-field test showed decreased habituation of Timp-3 KO mice. Immunostaining of brain slices revealed the expression of Timp-3 in the hippocampus. In situ zymography of the hippocampus showed increased gelatinolytic activity in Timp-3 KO mice compared with WT mice. These results present the first evidence of Timp-3 involvement in cognitive function and hippocampal MMP activity in mice. Moreover, our findings suggest a novel therapeutic target to be explored for improvement of cognitive function in humans.
Collapse
Affiliation(s)
- Yoshichika Baba
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Osamu Yasuda
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yukihiro Takemura
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasuyuki Ishikawa
- Division of Structural Cell Biology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, Nara, Japan
| | - Mitsuru Ohishi
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Jun Iwanami
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, Japan
| | - Masaki Mogi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, Japan
| | - Nobutaka Doe
- Section of Behavioral Science, Kouiken Co. Ltd., Akashi, Hyogo, Japan
| | - Masatsugu Horiuchi
- Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Shitsukawa, Tohon, Ehime, Japan
| | - Nobuyo Maeda
- Department of Pathology and Laboratory Medicine, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Keisuke Fukuo
- Department of Food Sciences and Nutrition, School of Human Environmental Sciences, Mukogawa Woman's University, Nishinomiya, Hyogo, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
48
|
Yang Y, Candelario-Jalil E, Thompson JF, Cuadrado E, Estrada EY, Rosell A, Montaner J, Rosenberg GA. Increased intranuclear matrix metalloproteinase activity in neurons interferes with oxidative DNA repair in focal cerebral ischemia. J Neurochem 2009; 112:134-49. [PMID: 19840223 DOI: 10.1111/j.1471-4159.2009.06433.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Increased matrix metalloproteinase (MMP) activity is implicated in proteolysis of extracellular matrix in ischemic stroke. We recently observed intranuclear MMP activity in ischemic brain neurons at early reperfusion, suggesting a possible role in nuclear matrix proteolysis. Nuclear proteins, poly-ADP-ribose polymerase-1 (PARP-1) and X-ray cross-complementary factor 1 (XRCC1), as well as DNA repair enzymes, are important in DNA fragmentation and cell apoptosis. We hypothesized that intranuclear MMP activity facilitates oxidative injury in neurons during early ischemic insult by cleaving PARP-1 and XRCC1, interfering with DNA repair. We induced a 90-min middle cerebral artery occlusion in rats. Increase activity of MMP-2 and -9, detected in the ischemic neuronal nuclei at 3 h, was associated with DNA fragmentation at 24 and 48 h reperfusion. The intranuclear MMPs cleaved PARP-1. Treatment of the rats with a broad-spectrum MMP inhibitor, BB1101, significantly attenuated ischemia-induced PARP-1 cleavage, increasing its activity. Degradation of XRCC1 caused by ischemic insult in rat brain was also significantly attenuated by BB1101. We found elevation of oxidized DNA, apurinic/apyrimidinic sites, and 8-hydroxy-2'-deoxyguanosine, in ischemic brain cells at 3 h reperfusion. BB1101 markedly attenuated the early increase of oxidized DNA. Using tissue from stroke patients, we found increased intranuclear MMP expression. Our data suggest that intranuclear MMP activity cleaves PARP-1 and XRCC1, interfering with oxidative DNA repair. This novel role for MMPs could contribute to neuronal apoptosis in ischemic injuries.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Barkalifa R, Hershfinkel M, Friedman JE, Kozak A, Sekler I. The lipophilic zinc chelator DP-b99 prevents zinc induced neuronal death. Eur J Pharmacol 2009; 618:15-21. [PMID: 19622352 DOI: 10.1016/j.ejphar.2009.07.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 06/29/2009] [Accepted: 07/09/2009] [Indexed: 10/20/2022]
Abstract
Zinc plays a key pathophysiological role in major neurological disorders as well as diabetes, while being essential for the activity of numerous zinc binding proteins. A major challenge in chelation based therapy must take into consideration these apparently conflicting effects of zinc. One approach is to limit the activity of the chelator to regions and levels of zinc pathology, making normal zinc-dependent processes invisible to the chelator. Combining fluorescent zinc imaging with cytotoxicity assays we studied the zinc chelation efficacy and neuroprotective effect of the lipophilic divalent transition metal chelator DP-b99 (1,2-Bis(2-amino-phenoxy)ethane-N,N,N',N'-tetraacetic acid-N-N'-di[2-(octyloxy)ethyl ester],-N,N'-disodium salt). The affinity of DP-b99 to Zn(2+) and Ca(2+) ions is moderate in water and enhanced significantly in the lipid milieu. Application of DP-b99 to MIN6 beta-cells that were preloaded with zinc was followed by a decrease in fluorescence of the intracellular Zn(2+) sensitive dye, ZnAF-2DA, to resting levels. Preloading of MIN6 cells with DP-b99 was also effective in attenuating subsequent cellular zinc rise. Concentration-dependence analysis of zinc accumulation indicated that DP-b99 acts as a zinc chelator with moderate affinity. DP-b99 preapplication attenuated both Zn(2+) and Ca(2+) rise in neuronal cultures and also Zn(2+) rise in brain slices. Finally, DP-b99 attenuated Zn(2+)-induced neuronal death. Our results indicate that DP-b99 is effective in attenuating Zn(2+) and Ca(2+) surges and protecting neurons against a toxic Zn(2+)-rise. This may underlie the efficacy of DP-b99 in stroke treatment.
Collapse
Affiliation(s)
- Ronit Barkalifa
- Morphology, and the Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | |
Collapse
|
50
|
Park KP, Rosell A, Foerch C, Xing C, Kim WJ, Lee S, Opdenakker G, Furie KL, Lo EH. Plasma and brain matrix metalloproteinase-9 after acute focal cerebral ischemia in rats. Stroke 2009; 40:2836-42. [PMID: 19556529 DOI: 10.1161/strokeaha.109.554824] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Plasma levels of matrix metalloproteinase-9 (MMP-9) have been proposed to be a useful biomarker for assessing pathological events in brain. Here, we examined the temporal profiles of MMP-9 in blood and brain using a rat model of acute focal cerebral ischemia. METHODS Plasma and brain levels of MMP-2 and MMP-9 were quantified at 3, 6, 12, and 24 hours after permanent middle cerebral artery occlusion in male Sprague-Dawley rats. Infarct volumes at 24 hours were confirmed with 2,3,5-triphenyl-tetrazolium-chloride staining. RESULTS In plasma, zymographic bands were detected between 70 and 95 kDa corresponding to pro-MMP-2, pro-MMP-9, and activated MMP-9. A higher 135-kDa band was also seen that is likely to be NGAL-conjugated MMP-9. After ischemia, there were no significant changes in pro-MMP-2, but plasma levels of pro-MMP-9 steadily increased over the course of 24 hours. Activated MMP-9 levels in plasma were significantly elevated only at 24 hours. Plasma NGAL-MMP-9 complexes showed a transient elevation between 3 to 6 hours, after which levels decreased back down to pre-ischemic baselines. In brain homogenates, pro-MMP-2, pro-MMP-9, and activated MMP-9 were seen but no NGAL-MMP-9 bands were detected. Compared to the contralateral hemisphere, MMP-2 and MMP-9 levels in ischemic brain progressively increased over the course of 24 hours. Overall levels of MMP-9 in plasma and brain were significantly correlated, especially at 24 hours. Plasma levels of pro-MMP-9 at 24 hours were correlated with final infarct volumes. CONCLUSIONS Elevated plasma levels of MMP-9 appear to be correlated with brain levels within 24 hours of acute cerebral ischemia in rats. Further investigation into clinical profiles of MMP-9 in acute stroke patients may be useful.
Collapse
Affiliation(s)
- Kyung-Pil Park
- Neuroprotection Research Laboratory, MGH East 149-2401 Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|