1
|
Slowikowski E, Willems C, Lemes RMR, Schuermans S, Berghmans N, Rocha RPF, Martens E, Proost P, Delang L, Marques RE, Filho JCA, Marques PE. A central role for CCR2 in monocyte recruitment and blood-brain barrier disruption during Usutu virus encephalitis. J Neuroinflammation 2025; 22:107. [PMID: 40241134 PMCID: PMC12004732 DOI: 10.1186/s12974-025-03435-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Usutu virus (USUV) is an emerging neurotropic flavivirus capable of causing encephalitis in humans. Here, our main goal was to characterize the innate immune response in the brain during USUV encephalitis and to identify strategies to control disease severity. Using an immunocompetent mouse model of USUV encephalitis, we showed that microglia activation, blood-brain barrier (BBB) disruption and inflammatory monocyte recruitment are hallmarks of disease 6 days post infection. Activated microglia were in close association to USUV-infected cells, concomitantly with elevated levels of IL-6, IFN-γ, CCL2, CCL5, CXCL10 and CXCL1 in the brain. Monocyte recruitment was CCR2-dependent and driven by IFN-γ and CCL2 production beneath the brain vasculature. Moreover, CCR2 deficiency inhibited microglia activation and BBB disruption, showing the central role of CCR2 in USUV encephalitis. Accordingly, treatment with dexamethasone prevented pro-inflammatory mediator production and reduced leukocyte recruitment significantly, restraining encephalitis severity. Concluding, USUV encephalitis is driven by CCR2-mediated monocyte recruitment and BBB disruption, and blocked therapeutically by glucocorticoids.
Collapse
Affiliation(s)
- Emily Slowikowski
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Céleste Willems
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Robertha Mariana Rodrigues Lemes
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sara Schuermans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Rebeca Paiva Fróes Rocha
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Erik Martens
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Delang
- Virus-Host Interactions and Therapeutic Approaches (VITA) Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - José Carlos Alves Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Sung W, Kim YS, Lee KY, Jung JA, Choi H, Lee YJ, Koh SH. Aspirin modulates inflammatory biomarkers in patients with subcortical silent brain infarcts. Front Aging Neurosci 2025; 16:1507683. [PMID: 39850789 PMCID: PMC11756530 DOI: 10.3389/fnagi.2024.1507683] [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: 10/08/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction This study aimed to identify differences in the levels of inflammation-related biomarkers between patients with subcortical silent brain infarcts (SBIs) and healthy controls. We also evaluated the effect of aspirin on the subcortical SBI inflammatory processes. Methods Consecutive patients diagnosed with subcortical SBIs without a history of acute stroke were included. The demographic and clinical data of the 26 subjects with subcortical SBIs, such as the number and location of subcortical SBIs, were reviewed. Plasma levels of macrophage migration inhibitory factor (MIF), matrix metalloproteinase-9 (MMP-9), and visfatin were measured in patients with subcortical SBIs and ten healthy participants. These biomarkers were rechecked in patients with subcortical SBI 3 months after taking aspirin (100 mg/day). Results MIF and MMP-9 levels were significantly higher in patients with subcortical SBIs than in healthy control group (p = 0.031 and p = 0.026, respectively). Although MIF and MMP-9 did not show significant changes after taking aspirin for 3 months, the median plasma level of visfatin was significantly decreased from 1.00 ng/mL (range, 0.86-1.16 ng/mL) to 0.84 ng/mL (range, 0.77-0.91 ng/mL) (p = 0.002) after taking aspirin. Discussion Inflammation could be an essential factor in the pathogenesis of subcortical SBIs, and aspirin affects several inflammation-related biomarkers.
Collapse
Affiliation(s)
- Wonjae Sung
- Department of Neurology, College of Medicine, Hanyang University Seoul Hospital, Seoul, Republic of Korea
| | - Young Seo Kim
- Department of Neurology, College of Medicine, Hanyang University Seoul Hospital, Seoul, Republic of Korea
| | - Kyu-Yong Lee
- Department of Neurology, College of Medicine, Hanyang University Guri Hospital, Guri, Republic of Korea
| | - Jae-A Jung
- Department of Plastic and Reconstructive Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Republic of Korea
| | - Hojin Choi
- Department of Neurology, College of Medicine, Hanyang University Guri Hospital, Guri, Republic of Korea
| | - Young Joo Lee
- Department of Neurology, College of Medicine, Hanyang University Guri Hospital, Guri, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, College of Medicine, Hanyang University Guri Hospital, Guri, Republic of Korea
- Department of Translational Medicine, Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Ganguly K, Adhikary K, Acharjee A, Acharjee P, Trigun SK, Mutlaq AS, Ashique S, Yasmin S, Alshahrani AM, Ansari MY. Biological significance and pathophysiological role of Matrix Metalloproteinases in the Central Nervous System. Int J Biol Macromol 2024; 280:135967. [PMID: 39322129 DOI: 10.1016/j.ijbiomac.2024.135967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/21/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Matrix Metalloproteinases (MMPs), which are endopeptidase reliant on zinc, are low in embryonic tissues but increases in response to a variety of physiological stimulus and pathological stresses. Neuro-glial cells, endothelial cells, fibroblasts, and leucocytes secrete MMPs, which cleave extracellular matrix proteins in a time-dependent manner. MMPs affect synaptic plasticity and the development of short-term memory by controlling the size, shape, and excitatory synapses' function through the lateral diffusion of receptors. In addition, MMPs influence the Extracellular Matrix proteins in the Peri-Neuronal Net at the Neuro-glial interface, which aids in the establishment of long-term memory. Through modulating neuronal, and glial cells migration, differentiation, Neurogenesis, and survival, MMPs impact brain development in mammals. In adult brains, MMPs play a beneficial role in physiological plasticity, which includes learning, memory consolidation, social interaction, and complex behaviors, by proteolytically altering a wide variety of factors, including growth factors, cytokines, receptors, DNA repair enzymes, and matrix proteins. Additionally, stress, depression, addiction, hepatic encephalopathy, and stroke may all have negative effects on MMPs. In addition to their role in glioblastoma development, MMPs influence neurological diseases such as epilepsy, schizophrenia, autism spectrum disorder, brain damage, pain, neurodegeneration, and Alzheimer's and Parkinson's. To help shed light on the potential of MMPs as a therapeutic target for neurodegenerative diseases, this review summarizes their regulation, mode of action, and participation in brain physiological plasticity and pathological damage. Finally, by employing different MMP-based nanotools and inhibitors, MMPs may also be utilized to map the anatomical and functional connectome of the brain, analyze its secretome, and treat neurodegenerative illnesses.
Collapse
Affiliation(s)
- Krishnendu Ganguly
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Krishnendu Adhikary
- Department of Medical Lab Technology, Paramedical College Durgapur, Helen Keller Sarani, Durgapur 713212, West Bengal, India.
| | - Arup Acharjee
- Molecular Omics Laboratory, Department of Zoology, University of Allahabad, Allahabad, Uttar Pradesh, India.
| | - Papia Acharjee
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | - Surendra Kumar Trigun
- Biochemistry Section, Department of Zoology, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
| | | | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India.
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia.
| | - Asma M Alshahrani
- Department of Clinical Pharmacy, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia; Department of Clinical Pharmacy, Shaqra University, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
4
|
Mishra RR, Nielsen BE, Trudrung MA, Lee S, Bolstad LJ, Hellenbrand DJ, Hanna AS. The Effect of Tissue Inhibitor of Metalloproteinases on Scar Formation after Spinal Cord Injury. Cells 2024; 13:1547. [PMID: 39329731 PMCID: PMC11430430 DOI: 10.3390/cells13181547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Spinal cord injury (SCI) often results in permanent loss of motor and sensory function. After SCI, the blood-spinal cord barrier (BSCB) is disrupted, causing the infiltration of neutrophils and macrophages, which secrete several kinds of cytokines, as well as matrix metalloproteinases (MMPs). MMPs are proteases capable of degrading various extracellular matrix (ECM) proteins, as well as many non-matrix substrates. The tissue inhibitor of MMPs (TIMP)-1 is significantly upregulated post-SCI and operates via MMP-dependent and MMP-independent pathways. Through the MMP-dependent pathway, TIMP-1 directly reduces inflammation and destruction of the ECM by binding and blocking the catalytic domains of MMPs. Thus, TIMP-1 helps preserve the BSCB and reduces immune cell infiltration. The MMP-independent pathway involves TIMP-1's cytokine-like functions, in which it binds specific TIMP surface receptors. Through receptor binding, TIMP-1 can stimulate the proliferation of several types of cells, including keratinocytes, aortic smooth muscle cells, skin epithelial cells, corneal epithelial cells, and astrocytes. TIMP-1 induces astrocyte proliferation, modulates microglia activation, and increases myelination and neurite extension in the central nervous system (CNS). In addition, TIMP-1 also regulates apoptosis and promotes cell survival through direct signaling. This review provides a comprehensive assessment of TIMP-1, specifically regarding its contribution to inflammation, ECM remodeling, and scar formation after SCI.
Collapse
Affiliation(s)
- Raveena R. Mishra
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Brooke E. Nielsen
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Melissa A. Trudrung
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Samuel Lee
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Luke J. Bolstad
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Daniel J. Hellenbrand
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amgad S. Hanna
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
5
|
Kreutz A, Oyetade OB, Chang X, Hsieh JH, Behl M, Allen DG, Kleinstreuer NC, Hogberg HT. Integrated Approach for Testing and Assessment for Developmental Neurotoxicity (DNT) to Prioritize Aromatic Organophosphorus Flame Retardants. TOXICS 2024; 12:437. [PMID: 38922117 PMCID: PMC11209292 DOI: 10.3390/toxics12060437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/27/2024]
Abstract
Organophosphorus flame retardants (OPFRs) are abundant and persistent in the environment but have limited toxicity information. Their similarity in structure to organophosphate pesticides presents great concern for developmental neurotoxicity (DNT). However, current in vivo testing is not suitable to provide DNT information on the amount of OPFRs that lack data. Over the past decade, an in vitro battery was developed to enhance DNT assessment, consisting of assays that evaluate cellular processes in neurodevelopment and function. In this study, behavioral data of small model organisms were also included. To assess if these assays provide sufficient mechanistic coverage to prioritize chemicals for further testing and/or identify hazards, an integrated approach to testing and assessment (IATA) was developed with additional information from the Integrated Chemical Environment (ICE) and the literature. Human biomonitoring and exposure data were identified and physiologically-based toxicokinetic models were applied to relate in vitro toxicity data to human exposure based on maximum plasma concentration. Eight OPFRs were evaluated, including aromatic OPFRs (triphenyl phosphate (TPHP), isopropylated phenyl phosphate (IPP), 2-ethylhexyl diphenyl phosphate (EHDP), tricresyl phosphate (TMPP), isodecyl diphenyl phosphate (IDDP), tert-butylphenyl diphenyl phosphate (BPDP)) and halogenated FRs ((Tris(1,3-dichloro-2-propyl) phosphate (TDCIPP), tris(2-chloroethyl) phosphate (TCEP)). Two representative brominated flame retardants (BFRs) (2,2'4,4'-tetrabromodiphenyl ether (BDE-47) and 3,3',5,5'-tetrabromobisphenol A (TBBPA)) with known DNT potential were selected for toxicity benchmarking. Data from the DNT battery indicate that the aromatic OPFRs have activity at similar concentrations as the BFRs and should therefore be evaluated further. However, these assays provide limited information on the mechanism of the compounds. By integrating information from ICE and the literature, endocrine disruption was identified as a potential mechanism. This IATA case study indicates that human exposure to some OPFRs could lead to a plasma concentration similar to those exerting in vitro activities, indicating potential concern for human health.
Collapse
Affiliation(s)
- Anna Kreutz
- Inotiv, Research Triangle Park, NC 27560, USA; (A.K.); (O.B.O.); (X.C.); (D.G.A.)
| | - Oluwakemi B. Oyetade
- Inotiv, Research Triangle Park, NC 27560, USA; (A.K.); (O.B.O.); (X.C.); (D.G.A.)
| | - Xiaoqing Chang
- Inotiv, Research Triangle Park, NC 27560, USA; (A.K.); (O.B.O.); (X.C.); (D.G.A.)
| | - Jui-Hua Hsieh
- NIH/NIEHS/DTT/PTB, Research Triangle Park, NC 27560, USA;
| | - Mamta Behl
- Neurocrine Biosciences Inc., San Diego, CA 92130, USA;
| | - David G. Allen
- Inotiv, Research Triangle Park, NC 27560, USA; (A.K.); (O.B.O.); (X.C.); (D.G.A.)
| | | | | |
Collapse
|
6
|
Lee TH, Chen JL, Chang CH, Tsai MM, Tseng HC, Chang YC, Shanmugam V, Hsieh HL. A Brain-Protective Sterol from Soft Coral Inhibits Lipopolysaccharide-Induced Matrix Metalloproteinase-9-Mediated Astrocytic Migration. Biomedicines 2024; 12:226. [PMID: 38275397 PMCID: PMC10813456 DOI: 10.3390/biomedicines12010226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Matrix metalloproteinases (MMPs), which are proteolytic enzymes, promote blood-brain barrier (BBB) disruption, leading to neuronal damage and neuroinflammation. Among them, MMP-9 upregulation serves as an inflammatory biomarker in the central nervous system (CNS). Currently, the development of marine organism-derived bioactive compounds or metabolites as anti-inflammatory drugs has received considerable attention. The 9,11-secosteroid, 3β,11-dihydroxy-9,11-secogorgost-5-en-9-one (4p3f), is a novel sterol compound extracted from the soft coral Sinularia leptoclado with potential anti-inflammatory activity. However, the effect of and potential for brain protection of 4p3f on brain astrocytes remain unclear. Herein, we used rat brain astrocytes (RBAs) to investigate the effects and signaling mechanisms of 4p3f on lipopolysaccharide (LPS)-induced MMP-9 expression via zymographic, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), Western blot, immunofluorescence staining, promoter-reporter, and cell migration analyses. We first found that 4p3f blocked LPS-induced MMP-9 expression in RBAs. Next, we demonstrated that LPS induced MMP-9 expression via the activation of ERK1/2, p38 MAPK, and JNK1/2, which is linked to the STAT3-mediated NF-κB signaling pathway. Finally, 4p3f effectively inhibited LPS-induced upregulation of MMP-9-triggered RBA cell migration. These data suggest that a novel sterol from soft coral, 4p3f, may have anti-inflammatory and brain-protective effects by attenuating these signaling pathways of MMP-9-mediated events in brain astrocytes. Accordingly, the soft coral-derived sterol 4p3f may emerge as a potential candidate for drug development or as a natural compound with neuroprotective properties.
Collapse
Affiliation(s)
- Tsong-Hai Lee
- Stroke Center and Stroke Section, Department of Neurology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Jiun-Liang Chen
- Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Chuan-Hsin Chang
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; (C.-H.C.); (M.-M.T.); (H.-C.T.); (Y.-C.C.)
| | - Ming-Ming Tsai
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; (C.-H.C.); (M.-M.T.); (H.-C.T.); (Y.-C.C.)
| | - Hui-Ching Tseng
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; (C.-H.C.); (M.-M.T.); (H.-C.T.); (Y.-C.C.)
| | - Yu-Chia Chang
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; (C.-H.C.); (M.-M.T.); (H.-C.T.); (Y.-C.C.)
| | | | - Hsi-Lung Hsieh
- Division of Basic Medical Sciences, Department of Nursing, Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan; (C.-H.C.); (M.-M.T.); (H.-C.T.); (Y.-C.C.)
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, R&D Center of Biochemical Engineering Technology, New Taipei City 301, Taiwan
| |
Collapse
|
7
|
Gao Y, Fang C, Wang J, Ye Y, Li Y, Xu Q, Kang X, Gu L. Neuroinflammatory Biomarkers in the Brain, Cerebrospinal Fluid, and Blood After Ischemic Stroke. Mol Neurobiol 2023; 60:5117-5136. [PMID: 37258724 DOI: 10.1007/s12035-023-03399-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
The most frequent type of stroke, known as ischemic stroke (IS), is a significant global public health issue. The pathological process of IS and post-IS episodes has not yet been fully explored, but neuroinflammation has been identified as one of the key processes. Biomarkers are objective indicators used to assess normal or pathological processes, evaluate responses to treatment, and predict outcomes, and some biomarkers can also be used as therapeutic targets. After IS, various molecules are produced by different cell types, such as microglia, astrocytes, infiltrating leukocytes, endothelial cells, and damaged neurons, that participate in the neuroinflammatory response within the ischemic brain region. These molecules may either promote or inhibit neuroinflammation and may be released into extracellular spaces, including cerebrospinal fluid (CSF) and blood, due to reasons such as BBB damage. These neuroinflammatory molecules should be valued as biomarkers to monitor whether their expression levels in the blood, CSF, and brain correlate with the diagnosis and prognosis of IS patients or whether they have potential as therapeutic targets. In addition, although some molecules do not directly participate in the process of neuroinflammation, they have been reported to have potential diagnostic or therapeutic value against post-IS neuroinflammation, and these molecules will also be listed. In this review, we summarize the neuroinflammatory biomarkers in the brain, CSF, and blood after an IS episode and the potential value of these biomarkers for the diagnosis, treatment, and prognosis of IS patients.
Collapse
Affiliation(s)
- Yikun Gao
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Congcong Fang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xianhui Kang
- Department of Anesthesia, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310006, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
8
|
Zhao J, Huh Y, Bortsov A, Diatchenko L, Ji RR. Immunotherapies in chronic pain through modulation of neuroimmune interactions. Pharmacol Ther 2023; 248:108476. [PMID: 37307899 PMCID: PMC10527194 DOI: 10.1016/j.pharmthera.2023.108476] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/18/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
It is generally believed that immune activation can elicit pain through production of inflammatory mediators that can activate nociceptive sensory neurons. Emerging evidence suggests that immune activation may also contribute to the resolution of pain by producing distinct pro-resolution/anti-inflammatory mediators. Recent research into the connection between the immune and nervous systems has opened new avenues for immunotherapy in pain management. This review provides an overview of the most utilized forms of immunotherapies (e.g., biologics) and highlight their potential for immune and neuronal modulation in chronic pain. Specifically, we discuss pain-related immunotherapy mechanisms that target inflammatory cytokine pathways, the PD-L1/PD-1 pathway, and the cGAS/STING pathway. This review also highlights cell-based immunotherapies targeting macrophages, T cells, neutrophils and mesenchymal stromal cells for chronic pain management.
Collapse
Affiliation(s)
- Junli Zhao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Andrey Bortsov
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC H3A 0G4, Canada; Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0G4, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
9
|
Liang E, Xiao S, Zhao C, Zhang Y, Fu G. M6A modification promotes blood-brain barrier breakdown during cerebral ischemia/reperfusion injury through increasing matrix metalloproteinase 3 expression. Heliyon 2023; 9:e16905. [PMID: 37332938 PMCID: PMC10275791 DOI: 10.1016/j.heliyon.2023.e16905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 04/26/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023] Open
Abstract
Blood-brain barrier (BBB) breakdown is a critical event in cerebral ischemia-reperfusion (I/R) injury, and matrix metalloproteinases (MMPs), which are proteolytic enzymes, play essential roles in BBB breakdown through degrading the extracellular matrix. N6-Methyladenosine (m6A), the most common and reversible mRNA modification, has an important role in the progression of cerebral I/R injury. However, whether m6A is related to BBB breakdown and MMPs expression in cerebral I/R injury is still not clear. In this study, we explored the potential effects of m6A modification on BBB breakdown in cerebral I/R injury and its underlying mechanisms using mice subjected to transient middle cerebral artery occlusion and reperfusion (MCAO/R), and mouse brain endothelial cells treated with oxygen-glucose deprivation and reoxygenation (OGD/R). We find that MMP3 expression is highly expressed and positively associated with the m6A writer CBLL1 (Cbl proto-oncogene like 1) in cerebral I/R injury in vivo and in vitro. Furthermore, MMP3 mRNA occurs m6A modification in mouse brain endothelial cells, and the m6A modification level of MMP3 mRNA is significantly increased in cerebral I/R injury. Moreover, inhibition of m6A modification reduces MMP3 expression and ameliorates BBB breakdown in cerebral I/R in vivo and in vitro. In conclusion, m6A modification promotes BBB breakdown in cerebral I/R injury through increasing MMP3 expression, indicating that m6A may be a potential therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- En Liang
- Department of Neurosurgery, Guangzhou PanYu He Xian Memorial Hospital, Guangzhou, 511442, PR China
| | - Shaorong Xiao
- Centre for Integrative Medicine, School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Changtong Zhao
- Department of Neurosurgery, Guangzhou PanYu He Xian Memorial Hospital, Guangzhou, 511442, PR China
| | - Yu Zhang
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518133, PR China
| | - Guanglei Fu
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, PR China
| |
Collapse
|
10
|
Lukito PP, July J, Suntoro VA, Wijaya JH, Hamdoyo A, Sindunata NA, Muljadi R. Neutrophil-to-lymphocyte ratio predicted cerebral infarction and poor discharge functional outcome in aneurysmal subarachnoid hemorrhage: A propensity score matching analysis. Surg Neurol Int 2023; 14:182. [PMID: 37292403 PMCID: PMC10246379 DOI: 10.25259/sni_127_2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/03/2023] [Indexed: 06/10/2023] Open
Abstract
Background Neutrophil-lymphocyte-ratio (NLR) and platelet-lymphocyte-ratio (PLR) have emerged as potential biomarkers in predicting the outcomes of aneurysmal subarachnoid hemorrhage (aSAH). Since a study was never conducted on the Southeast Asian and Indonesian population, we designed the present study to evaluate the potential of NLR and PLR in predicting cerebral infarction and functional outcomes and find the optimal cutoff value. Methods We retrospectively reviewed patients admitted for aSAH in our hospital between 2017 and 2021. The diagnosis was made using a computed tomography (CT) scan or magnetic resonance imaging and CT angiography. Association between admission NLR and PLR and the outcomes were analyzed using a multivariable regression model. A receiver operating characteristic (ROC) analysis was done to identify the optimal cutoff value. A propensity score matching (PSM) was then carried out to reduce the imbalance between the two groups before comparison. Results Sixty-three patients were included in the study. NLR was independently associated with cerebral infarction (odds ratio, OR 1.197 [95% confidence interval, CI 1.027-1.395] per 1-point increment; P = 0.021) and poor discharge functional outcome (OR 1.175 [95% CI 1.036-1.334] per 1-point increment; P = 0.012). PLR did not significantly correlate with the outcomes. ROC analysis identified 7.09 as the cutoff for cerebral infarction and 7.50 for discharge functional outcome. Dichotomizing and performing PSM revealed that patients with NLR above the identified cutoff value significantly had more cerebral infarction and poor discharge functional outcome. Conclusion NLR demonstrated a good prognostic capability in Indonesian aSAH patients. More studies should be conducted to find the optimal cutoff value for each population.
Collapse
Affiliation(s)
- Patrick Putra Lukito
- Department of Neurosurgery, Neuroscience Center Siloam Hospital, Tangerang, Banten, Indonesia
| | - Julius July
- Department of Neurosurgery, Neuroscience Center Siloam Hospital, Tangerang, Banten, Indonesia
| | | | - Jeremiah Hilkiah Wijaya
- Department of Neurosurgery, Neuroscience Center Siloam Hospital, Tangerang, Banten, Indonesia
| | - Audrey Hamdoyo
- Department of Neurosurgery, Neuroscience Center Siloam Hospital, Tangerang, Banten, Indonesia
| | - Nyoman Aditya Sindunata
- Department of Radiology, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Banten, Indonesia
| | - Rusli Muljadi
- Department of Radiology, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Banten, Indonesia
| |
Collapse
|
11
|
Zhang J, Li Y, Zhou T. Nerolidol Attenuates Cerebral Ischemic Injury in Middle Cerebral Artery Occlusion-Induced Rats via Regulation of Inflammation, Apoptosis, and Oxidative Stress Markers. Pharmacogn Mag 2023. [DOI: 10.1177/09731296221137380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023] Open
Abstract
Background Cerebral ischemia is a syndrome that occurs due to the restricted flow of oxygen-rich blood to the brain, causing damage to the brain cells. Globally, ischemia ranks second in causing mortality and third in causing disability in stroke patients. Nerolidol is a bioactive compound present in the essential oil of plants with a floral odour. It is a natural sesquiterpene alcohol used in cosmetics, perfumes, and as a food flavouring agent. It also possesses antioxidant, antimicrobial, anti-inflammatory, and anticancer properties. Materials and Methods In this study, we assessed the anti-ischemic property of nerolidol in cerebral ischemia-induced mice. Healthy male Wistar rats were induced into cerebral ischemia with middle cerebral artery occlusion (MCAO) and treated with 10 mg and 20 mg nerolidol for 21 days. The brain morphometric, antioxidant, and MMP levels were estimated in the brain tissue of MCAO-performed and nerolidol-treated rats. The cerebral infarct-alleviating potency of nerolidol was analysed by estimating the levels of inflammatory cytokines and apoptotic proteins. It was further confirmed by assessing the levels of COX-2/PGE-2 signalling proteins in brain tissue from MCAO-performed in rats. Results Nerolidol significantly reduced the cerebral infarct volume and brain edema via increased antioxidant levels and decreased MMPs. It also decreased the pro-inflammatory cytokines and proapoptotic proteins in brain tissue. The inflammatory signalling proteins NFκB, COX-2, and PGE-2 were significantly decreased in nerolidol-treated MCAO-performed rats, confirming the antiischemic property of nerolidol. Conclusion Our results prove nerolidol significantly alleviates cerebral ischemia in rats, and it can be subjected to further trials to be formulated as an anti-ischemic drug.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurosurgery, Laizhou City People’s Hospital, Laizhou, Shandong, China
| | - Yanli Li
- School of Health, Binzhou Polytechnical College, Binzhou, Shandong, China
| | - Tao Zhou
- Department of Neurosurgery, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
12
|
Thapa K, Shivam K, Khan H, Kaur A, Dua K, Singh S, Singh TG. Emerging Targets for Modulation of Immune Response and Inflammation in Stroke. Neurochem Res 2023; 48:1663-1690. [PMID: 36763312 DOI: 10.1007/s11064-023-03875-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023]
Abstract
The inflammatory and immunological responses play a significant role after stroke. The innate immune activation stimulated by microglia during stroke results in the migration of macrophages and lymphocytes into the brain and are responsible for tissue damage. The immune response and inflammation following stroke have no defined targets, and the intricacies of the immunological and inflammatory processes are only partially understood. Innate immune cells enter the brain and meninges during the acute phase, which can cause ischemia damage. Activation of systemic immunity is caused by danger signals sent into the bloodstream by injured brain cells, which is followed by a significant immunodepression that encourages life-threatening infections. Neuropsychiatric sequelae, a major source of post-stroke morbidity, may be induced by an adaptive immune response that is initiated by antigen presentation during the chronic period and is directed against the brain. Thus, the current review discusses the role of immune response and inflammation in stroke pathogenesis, their role in the progression of injury during the stroke, and the emerging targets for the modulation of the mechanism of immune response and inflammation that may have possible therapeutic benefits against stroke.
Collapse
Affiliation(s)
- Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.,School of Pharmacy, Chitkara University, Rajpura, Himachal Pradesh, 174103, India
| | - Kumar Shivam
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, 2007, Australia.,Faculty of Health, Australian Research Centre in Complementary & Integrative Medicine, University of Technology Sydney, Ultimo, 2007, Australia
| | - Sachin Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
13
|
Xie M, Hao Y, Feng L, Wang T, Yao M, Li H, Ma D, Feng J. Neutrophil Heterogeneity and its Roles in the Inflammatory Network after Ischemic Stroke. Curr Neuropharmacol 2023; 21:621-650. [PMID: 35794770 PMCID: PMC10207908 DOI: 10.2174/1570159x20666220706115957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
As the first peripheral immune cells to enter the brain after ischemic stroke, neutrophils are important participants in stroke-related neuroinflammation. Neutrophils are quickly mobilized from the periphery in response to a stroke episode and cross the blood-brain barrier to reach the ischemic brain parenchyma. This process involves the mobilization and activation of neutrophils from peripheral immune organs (including the bone marrow and spleen), their chemotaxis in the peripheral blood, and their infiltration into the brain parenchyma (including disruption of the blood-brain barrier, inflammatory effects on brain tissue, and interactions with other immune cell types). In the past, it was believed that neutrophils aggravated brain injuries through the massive release of proteases, reactive oxygen species, pro-inflammatory factors, and extracellular structures known as neutrophil extracellular traps (NETs). With the failure of early clinical trials targeting neutrophils and uncovering their underlying heterogeneity, our view of their role in ischemic stroke has become more complex and multifaceted. As neutrophils can be divided into N1 and N2 phenotypes in tumors, neutrophils have also been found to have similar phenotypes after ischemic stroke, and play different roles in the development and prognosis of ischemic stroke. N1 neutrophils are dominant during the acute phase of stroke (within three days) and are responsible for the damage to neural structures via the aforementioned mechanisms. However, the proportion of N2 neutrophils gradually increases in later phases, and this has a beneficial effect through the release of anti-inflammatory factors and other neuroprotective mediators. Moreover, the N1 and N2 phenotypes are highly plastic and can be transformed into each other under certain conditions. The pronounced differences in their function and their high degree of plasticity make these neutrophil subpopulations promising targets for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Meizhen Xie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Liangshu Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Tian Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Mengyue Yao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin, Changchun, Jilin Province 130021, China
| |
Collapse
|
14
|
Buchheit T. Platelet-Rich Plasma and Autologous Conditioned Serum: Non-Cellular Biologic Therapies for Neuroimmune Modulation and the Treatment of Arthritis Pain. NEUROIMMUNE INTERACTIONS IN PAIN 2023:287-303. [DOI: 10.1007/978-3-031-29231-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Khotimchenko YS, Silachev DN, Katanaev VL. Marine Natural Products from the Russian Pacific as Sources of Drugs for Neurodegenerative Diseases. Mar Drugs 2022; 20:708. [PMID: 36421986 PMCID: PMC9697637 DOI: 10.3390/md20110708] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 09/05/2023] Open
Abstract
Neurodegenerative diseases are growing to become one of humanity's biggest health problems, given the number of individuals affected by them. They cause enough mortalities and severe economic impact to rival cancers and infections. With the current diversity of pathophysiological mechanisms involved in neurodegenerative diseases, on the one hand, and scarcity of efficient prevention and treatment strategies, on the other, all possible sources for novel drug discovery must be employed. Marine pharmacology represents a relatively uncharted territory to seek promising compounds, despite the enormous chemodiversity it offers. The current work discusses one vast marine region-the Northwestern or Russian Pacific-as the treasure chest for marine-based drug discovery targeting neurodegenerative diseases. We overview the natural products of neurological properties already discovered from its waters and survey the existing molecular and cellular targets for pharmacological modulation of the disease. We further provide a general assessment of the drug discovery potential of the Russian Pacific in case of its systematic development to tackle neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuri S. Khotimchenko
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 8 ul. Sukhanova, 690950 Vladivostok, Russia
- A.V. Zhirmunsky National Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690950 Vladivostok, Russia
| | - Denis N. Silachev
- Department of Functional Biochemistry of Biopolymers, A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Vladimir L. Katanaev
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 8 ul. Sukhanova, 690950 Vladivostok, Russia
- Department of Cell Physiology and Metabolism, Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| |
Collapse
|
16
|
Tsiknia AA, Sundermann EE, Reas ET, Edland SD, Brewer JB, Galasko D, Banks SJ. Sex differences in Alzheimer's disease: plasma MMP-9 and markers of disease severity. Alzheimers Res Ther 2022; 14:160. [PMID: 36324151 PMCID: PMC9628176 DOI: 10.1186/s13195-022-01106-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/16/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Studies have reported higher plasma matrix metalloproteinase-9 (MMP-9) levels in mild cognitive impairment (MCI) and Alzheimer's disease (AD). Despite evidence that MMP-9 activity and its influence on AD pathophysiology may be modulated by sex hormones, sex differences in the association between MMP-9 and AD biomarkers and cognition have not been explored. METHODS Our sample included 238 amyloid-β (Aβ)-positive participants with MCI or AD dementia from the Alzheimer's Disease Neuroimaging Initiative (37.4% women, 74.6 ± 7.3 years). We used linear regression models to examine whether sex modified free and total plasma MMP-9 associations with CSF t-tau, p-tau181, and Aβ42. We used linear mixed effects models to examine whether sex modified total and free plasma MMP-9 associations with cognition, using longitudinal Mini-Mental Status Examination (MMSE) and Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-cog) data. RESULTS Total and free MMP-9 levels did not differ by sex, but AD dementia patients had higher total MMP-9 levels than participants with MCI (β = 0.06 [-0.11 to -0.01], p = 0.031). Sex modified the association of CSF t-tau with total (β = 128.68 [55.37 to 201.99], p < 0.001) and free MMP-9 (β = 98.61 [33.61 to 163.62], p = 0.003), whereby higher total and free MMP-9 correlated with higher CSF t-tau in women and lower CSF t-tau in men. Higher free MMP-9 correlated with lower CSF p-tau181 among men (β = -14.98 [-27.37 to -2.58], p = 0.018), but not women. In participants with MCI, higher free MMP-9 levels were associated with higher CSF Aβ42 among men (β = 26.88 [4.03 to 49.73], p = 0.022) but not women. In the overall sample, higher free and total MMP-9 at baseline predicted worsening MMSE scores in women (β = -2.10 [-3.97 to -0.27], p = 0.027 and β = -2.24 [-4.32 to -0.18], p = 0.035) but not men. Higher free MMP-9 correlated with worse ADAS-cog scores (β = 12.34 [3.02 to 21.65], p = 0.011) in women (β = 12.34 [3.02 to 21.65], p = 0.011) but not men with AD dementia cross-sectionally but correlated with worsening ADAS-cog scores longitudinally only in men (β = 8.98 [0.27 to 17.68], p = 0.042). CONCLUSIONS MMP-9 may have more detrimental effects on AD-related pathological and cognitive changes in women. If replicated, our findings could help uncover potential mechanisms contributing to women's elevated susceptibility to AD.
Collapse
Affiliation(s)
- Amaryllis A. Tsiknia
- Department of Neurosciences, School of Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093 USA
| | - Erin E. Sundermann
- grid.410371.00000 0004 0419 2708Research Service, VA San Diego Healthcare System, San Diego, CA 92161 USA ,grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093 USA
| | - Emilie T. Reas
- Department of Neurosciences, School of Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093 USA
| | - Steven D. Edland
- Department of Neurosciences, School of Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093 USA ,grid.266100.30000 0001 2107 4242Division of Biostatistics, School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA 92093 USA
| | - James B. Brewer
- Department of Neurosciences, School of Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093 USA
| | - Douglas Galasko
- Department of Neurosciences, School of Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093 USA
| | - Sarah J. Banks
- Department of Neurosciences, School of Medicine, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093 USA
| | | |
Collapse
|
17
|
Cai H, Huang LY, Hong R, Song JX, Guo XJ, Zhou W, Hu ZL, Wang W, Wang YL, Shen JG, Qi SH. Momordica charantia Exosome-Like Nanoparticles Exert Neuroprotective Effects Against Ischemic Brain Injury via Inhibiting Matrix Metalloproteinase 9 and Activating the AKT/GSK3β Signaling Pathway. Front Pharmacol 2022; 13:908830. [PMID: 35814200 PMCID: PMC9263912 DOI: 10.3389/fphar.2022.908830] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Plant exosome-like nanoparticles (ELNs) have shown great potential in treating tumor and inflammatory diseases, but the neuroprotective effect of plant ELNs remains unknown. In the present study, we isolated and characterized novel ELNs from Momordica charantia (MC) and investigated their neuroprotective effects against cerebral ischemia-reperfusion injury. In the present study, MC-ELNs were isolated by ultracentrifugation and characterized. Male Sprague–Dawley rats were subjected to middle cerebral artery occlusion (MCAO) and MC-ELN injection intravenously. The integrity of the blood–brain barrier (BBB) was examined by Evans blue staining and with the expression of matrix metalloproteinase 9 (MMP-9), claudin-5, and ZO-1. Neuronal apoptosis was evaluated by TUNEL and the expression of apoptotic proteins including Bcl2, Bax, and cleaved caspase 3. The major discoveries include: 1) Dil-labeled MC-ELNs were identified in the infarct area; 2) MC-ELN treatment significantly ameliorated BBB disruption, decreased infarct sizes, and reduced neurological deficit scores; 3) MC-ELN treatment obviously downregulated the expression of MMP-9 and upregulated the expression of ZO-1 and claudin-5. Small RNA-sequencing revealed that MC-ELN-derived miRNA5266 reduced MMP-9 expression. Furthermore, MC-ELN treatment significantly upregulated the AKT/GSK3β signaling pathway and attenuated neuronal apoptosis in HT22 cells. Taken together, these findings indicate that MC-ELNs attenuate ischemia-reperfusion–induced damage to the BBB and inhibit neuronal apoptosis probably via the upregulation of the AKT/GSK3β signaling pathway.
Collapse
Affiliation(s)
- Heng Cai
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Lin-Yan Huang
- Medical and Technology School, Xuzhou Medical University, And Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
| | - Rui Hong
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Jin-Xiu Song
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
| | - Xin-Jian Guo
- Medical and Technology School, Xuzhou Medical University, And Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
| | - Wei Zhou
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Zhao-Li Hu
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, China
| | - Wan Wang
- Medical and Technology School, Xuzhou Medical University, And Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
| | - Yan-Ling Wang
- Medical and Technology School, Xuzhou Medical University, And Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
| | - Jian-Gang Shen
- Medical and Technology School, Xuzhou Medical University, And Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
- *Correspondence: Su-Hua Qi, ; Jian-Gang Shen,
| | - Su-Hua Qi
- Pharmacology College, Xuzhou Medical University, Xuzhou, China
- Medical and Technology School, Xuzhou Medical University, And Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou, China
- *Correspondence: Su-Hua Qi, ; Jian-Gang Shen,
| |
Collapse
|
18
|
Grape-Seed-Derived Procyanidin Attenuates Chemotherapy-Induced Cognitive Impairment by Suppressing MMP-9 Activity and Related Blood–Brain-Barrier Damage. Brain Sci 2022; 12:brainsci12050571. [PMID: 35624958 PMCID: PMC9139059 DOI: 10.3390/brainsci12050571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Chemotherapy-induced cognitive impairment (CICI) is often observed in cancer patients and impairs their life quality. Grape-seed-orientated procyanidin has been shown to have anti-inflammatory and neuroprotective effects, yet its effects in preventing CICI have not been investigated. (2) Method: Adult male mice received 2.3 mg/kg cisplatin or saline injections for three cycles consisting of five daily injections followed by 5 days of rest. Procyanidin or saline was administered 1 h prior to cisplatin treatment. Cognitive testing, gelatin zymography, and blood–brain-barrier (BBB) penetration tests were performed after treatment cessation. RAW264.7 cells were treated by stimulated supernatant of SHSY5Y cells. In addition, high-mobility group protein B1 (HMGB1) expression and MMP-9 activity were tested. (3) Results: Repeated cisplatin treatment increased BBB penetration, MMP-9 activity, impaired performance in contextual fear conditioning, and novel object recognition tasks. The knockout of MMP-9 rescues cognitive impairment and cisplatin-induced upregulation of HMGB1 in SHSY5Y cells. HMGB1/TLR4/IP3K/AKT signaling contributes to the increased MMP-9 activity in RAW264.7 cells. Procyanidin treatment attenuates MMP-9 activity, BBB damage, and CICI. (4) Conclusions: The results indicated that MMP-9 activation and BBB disruption is involved in CICI. Procyanidin may effectively alleviate the harmful effects of cisplatin.
Collapse
|
19
|
Robinson BD, Isbell CL, Melge AR, Lomas AM, Shaji CA, Mohan CG, Huang JH, Tharakan B. Doxycycline prevents blood-brain barrier dysfunction and microvascular hyperpermeability after traumatic brain injury. Sci Rep 2022; 12:5415. [PMID: 35354869 PMCID: PMC8967830 DOI: 10.1038/s41598-022-09394-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/09/2022] [Indexed: 11/30/2022] Open
Abstract
The main objective of this study was to determine the cellular and molecular effects of doxycycline on the blood–brain barrier (BBB) and protection against secondary injuries following traumatic brain injury (TBI). Microvascular hyperpermeability and cerebral edema resulting from BBB dysfunction after TBI leads to elevation of intracranial pressure, secondary brain ischemia, herniation, and brain death. There are currently no effective therapies to modulate the underlying pathophysiology responsible for TBI-induced BBB dysfunction and hyperpermeability. The loss of BBB integrity by the proteolytic enzyme matrix metalloproteinase-9 (MMP-9) is critical to TBI-induced BBB hyperpermeability, and doxycycline possesses anti-MMP-9 effect. In this study, the effect of doxycycline on BBB hyperpermeability was studied utilizing molecular modeling (using Glide) in silico, cell culture-based models in vitro, and a mouse model of TBI in vivo. Brain microvascular endothelial cell assays of tight junction protein immunofluorescence and barrier permeability were performed. Adult C57BL/6 mice were subjected to sham versus TBI with or without doxycycline treatment and immediate intravital microscopic analysis for evaluating BBB integrity. Postmortem mouse brain tissue was collected to measure MMP-9 enzyme activity. It was found that doxycycline binding to the MMP-9 active sites have binding affinity of −7.07 kcal/mol. Doxycycline treated cell monolayers were protected from microvascular hyperpermeability and retained tight junction integrity (p < 0.05). Doxycycline treatment decreased BBB hyperpermeability following TBI in mice by 25% (p < 0.05). MMP-9 enzyme activity in brain tissue decreased with doxycycline treatment following TBI (p < 0.05). Doxycycline preserves BBB tight junction integrity following TBI via inhibiting MMP-9 activity. When established in human subjects, doxycycline, may provide readily accessible medical treatment after TBI to attenuate secondary injury.
Collapse
Affiliation(s)
- Bobby D Robinson
- Department of Surgery, Baylor Scott and White Medical Center, Baylor Scott and White Research Institute, Temple, TX, USA.,Texas A&M University Health Science Center College of Medicine, Temple, TX, USA
| | - Claire L Isbell
- Department of Surgery, Baylor Scott and White Medical Center, Baylor Scott and White Research Institute, Temple, TX, USA.,Texas A&M University Health Science Center College of Medicine, Temple, TX, USA
| | - Anu R Melge
- Amrita Center for Nanosciences and Molecular Medicine, Kochi, Kerala, India
| | - Angela M Lomas
- Department of Surgery, Baylor Scott and White Medical Center, Baylor Scott and White Research Institute, Temple, TX, USA.,Texas A&M University Health Science Center College of Medicine, Temple, TX, USA
| | - Chinchusha Anasooya Shaji
- Department of Surgery, Baylor Scott and White Medical Center, Baylor Scott and White Research Institute, Temple, TX, USA
| | - C Gopi Mohan
- Amrita Center for Nanosciences and Molecular Medicine, Kochi, Kerala, India
| | - Jason H Huang
- Department of Neurosurgery, Texas A&M University Health Science Center College of Medicine, Temple, TX, USA
| | - Binu Tharakan
- Department of Surgery, Baylor Scott and White Medical Center, Baylor Scott and White Research Institute, Temple, TX, USA. .,Texas A&M University Health Science Center College of Medicine, Temple, TX, USA. .,Department of Surgery, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA.
| |
Collapse
|
20
|
Onwuha‐Ekpete L, Fields GB. Application of a triple‐helical peptide inhibitor of
MMP
‐2/
MMP
‐9 to examine T‐cell activation in experimental autoimmune encephalomyelitis. Pept Sci (Hoboken) 2022. [DOI: 10.1002/pep2.24262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Lillian Onwuha‐Ekpete
- The Institute for Human Health & Disease Intervention (I‐HEALTH) Florida Atlantic University Jupiter Florida USA
| | - Gregg B. Fields
- The Institute for Human Health & Disease Intervention (I‐HEALTH) Florida Atlantic University Jupiter Florida USA
- Department of Chemistry The Scripps Research Institute/Scripps Florida Jupiter Florida USA
| |
Collapse
|
21
|
Rahman MM, Islam MR, Islam MT, Harun-Or-Rashid M, Islam M, Abdullah S, Uddin MB, Das S, Rahaman MS, Ahmed M, Alhumaydhi FA, Emran TB, Mohamed AAR, Faruque MRI, Khandaker MU, Mostafa-Hedeab G. Stem Cell Transplantation Therapy and Neurological Disorders: Current Status and Future Perspectives. BIOLOGY 2022; 11:147. [PMID: 35053145 PMCID: PMC8772847 DOI: 10.3390/biology11010147] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases are a global health issue with inadequate therapeutic options and an inability to restore the damaged nervous system. With advances in technology, health scientists continue to identify new approaches to the treatment of neurodegenerative diseases. Lost or injured neurons and glial cells can lead to the development of several neurological diseases, including Parkinson's disease, stroke, and multiple sclerosis. In recent years, neurons and glial cells have successfully been generated from stem cells in the laboratory utilizing cell culture technologies, fueling efforts to develop stem cell-based transplantation therapies for human patients. When a stem cell divides, each new cell has the potential to either remain a stem cell or differentiate into a germ cell with specialized characteristics, such as muscle cells, red blood cells, or brain cells. Although several obstacles remain before stem cells can be used for clinical applications, including some potential disadvantages that must be overcome, this cellular development represents a potential pathway through which patients may eventually achieve the ability to live more normal lives. In this review, we summarize the stem cell-based therapies that have been explored for various neurological disorders, discuss the potential advantages and drawbacks of these therapies, and examine future directions for this field.
Collapse
Affiliation(s)
- Mohammad Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Touhidul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Harun-Or-Rashid
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mahfuzul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Sabirin Abdullah
- Space Science Center, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Mohammad Borhan Uddin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Sumit Das
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Mohammad Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.M.R.); (M.R.I.); (M.T.I.); (M.H.-O.-R.); (M.I.); (M.B.U.); (S.D.); (M.S.R.); (M.A.)
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | | | | | - Mayeen Uddin Khandaker
- Centre for Applied Physics and Radiation Technologies, School of Engineering and Technology, Sunway University, Bandar Sunway 47500, Selangor, Malaysia;
| | - Gomaa Mostafa-Hedeab
- Pharmacology Department & Health Sciences Research Unit, Medical College, Jouf University, Sakaka 72446, Saudi Arabia;
- Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef 62521, Egypt
| |
Collapse
|
22
|
Jiang B, Zhang Y, Wang Y, Li Z, Chen Q, Tang J, Zhu G. Glibenclamide Attenuates Neuroinflammation and Promotes Neurological Recovery After Intracerebral Hemorrhage in Aged Rats. Front Aging Neurosci 2021; 13:729652. [PMID: 34512312 PMCID: PMC8427510 DOI: 10.3389/fnagi.2021.729652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/21/2021] [Indexed: 11/13/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a common disease in the elderly population. Inflammation following ICH plays a detrimental role in secondary brain injury, which is associated with a poor prognosis of patients with ICH, and no efficient pharmacological preventions are available. Here, we investigated the effects of glibenclamide (GLC) on neuroinflammation in an autoblood-induced aged rat (18 months old) model of ICH. Rats were randomized into the sham, vehicle, and GLC groups. First, we investigated the expression level of sulfonylurea receptor 1 (Sur1) surrounding the hematoma after ICH. Then, neurological scores were calculated, and water maze tests, brain water content analysis, western blotting, and immunofluorescence assays were implemented to detect the neuroprotective effect of GLC. The expression of the Sur1-Trpm4 channel was significantly increased in the perihematomal tissue following ICH in aged rats. The GLC administration effectively reduced brain edema and improved neurofunction deficits following ICH. In addition, GLC increased the expression of brain-derived neurotrophic factors and decreased the expression of proinflammatory factors [tumor necrosis factor (TNF)-α,interleukin (IL)-1, and IL-6]. Moreover, GLC markedly reduced Ikappa-B (IκB) kinase (IKK) expression in microglia and nuclear factor (NF)-κB-P65 levels in perihematomal tissue. GLC ameliorated ICH-induced neuroinflammation and improved neurological outcomes in aged rats. In part, GLC may exert these effects by regulating the NF-κB signaling pathway through the Sur1-Trpm4 channel.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Neurology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Ying Zhang
- Department of Neurology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Yan Wang
- Department of Neurology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Zheng Li
- Department of Neurology, Chengdu Fifth People's Hospital, Chengdu, China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Jun Tang
- Department of Neurosurgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Charzewski Ł, Krzyśko KA, Lesyng B. Structural characterisation of inhibitory and non-inhibitory MMP-9-TIMP-1 complexes and implications for regulatory mechanisms of MMP-9. Sci Rep 2021; 11:13376. [PMID: 34183752 PMCID: PMC8238946 DOI: 10.1038/s41598-021-92881-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 05/12/2021] [Indexed: 12/18/2022] Open
Abstract
MMP-9 plays a number of important physiological functions but is also responsible for many pathological processes, including cancer invasion, metastasis, and angiogenesis. It is, therefore, crucial to understand its enzymatic activity, including activation and inhibition mechanisms. This enzyme may also be partially involved in the "cytokine storm" that is characteristic of COVID-19 disease (SARS-CoV-2), as well as in the molecular mechanisms responsible for lung fibrosis. Due to the variety of processing pathways involving MMP-9 in biological systems and its uniqueness due to the O-glycosylated domain (OGD) and fibronectin-like (FBN) domain, specific interactions with its natural TIMP-1 inhibitor should be carefully studied, because they differ significantly from other homologous systems. In particular, earlier experimental studies have indicated that the newly characterised circular form of a proMMP-9 homotrimer exhibits stronger binding properties to TIMP-1 compared to its monomeric form. However, molecular structures of the complexes and the binding mechanisms remain unknown. The purpose of this study is to fill in the gaps in knowledge. Molecular modelling methods are applied to build the inhibitory and non-inhibitory MMP-9-TIMP-1 complexes, which allows for a detailed description of these structures and should allow for a better understanding of the regulatory processes in which MMP-9 is involved.
Collapse
Affiliation(s)
- Łukasz Charzewski
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| | - Krystiana A Krzyśko
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland.
| | - Bogdan Lesyng
- Department of Biophysics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093, Warsaw, Poland
| |
Collapse
|
24
|
Wu S, Guo T, Qi W, Li Y, Gu J, Liu C, Sha Y, Yang B, Hu S, Zong X. Curcumin ameliorates ischemic stroke injury in rats by protecting the integrity of the blood-brain barrier. Exp Ther Med 2021; 22:783. [PMID: 34055082 PMCID: PMC8145684 DOI: 10.3892/etm.2021.10215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/16/2021] [Indexed: 12/16/2022] Open
Abstract
The blood-brain barrier (BBB) is critical for proper cerebral homeostasis and its dysfunction during ischemic stroke can result in significant neurological injury. The major goal of the present study was to identify whether curcumin pretreatment possessed protective effects on BBB integrity during the 24 h of acute ischemic brain injury. To investigate the protective effects of curcumin, male Sprague-Dawley rats were divided into multiple groups, including sham, middle cerebral artery occlusion/reperfusion (MCAO/R) vehicle and curcumin pretreated MCAO/R groups. The effects of curcumin were measured by analyzing neurological deficits, infarct size, BBB permeability and expression levels of permeability-related proteins in the brain. It was found that curcumin pretreatment significantly improved neurological scores, decreased infarct size, and protected synaptic remodeling of hippocampal neurons and upregulated the protein expression level of tight junction proteins, ZO-1, occludin and claudin-5 in ischemic rat brains. Furthermore, curcumin pretreatment before stroke was shown to downregulate the phosphorylation of NF-κB and MMP-9, which are central mediators of inflammation. The results from the present study indicated that curcumin pretreatment ameliorated ischemic stroke injury by protecting BBB integrity and synaptic remodeling, as well as inhibiting inflammatory responses.
Collapse
Affiliation(s)
- Shuguang Wu
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 313000, P.R. China
| | - Ting Guo
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Wenxuan Qi
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yuyu Li
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Jie Gu
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Cui Liu
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yuehong Sha
- Department of Emergency, First People's Hospital, Pizhou, Jiangsu 221300, P.R. China
| | - Baocheng Yang
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Emergency Center of The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Shuqun Hu
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Emergency Center of The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xuemei Zong
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Emergency Center of The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
25
|
Hypermethylation of EFEMP1 in the Hippocampus May Be Related to the Deficit in Spatial Memory of Rat Neonates Triggered by Repeated Administration of Propofol. BIOMED RESEARCH INTERNATIONAL 2021; 2020:8851480. [PMID: 33381589 PMCID: PMC7765714 DOI: 10.1155/2020/8851480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/22/2020] [Accepted: 11/30/2020] [Indexed: 11/17/2022]
Abstract
It has been confirmed that repeated application of propofol, as an intravenous and short-fast-acting anesthetic, in neonatal animals or humans may produce long-term deficits in cognitive functions. With the aim of explaining the neurotoxic effects of repeated administration of propofol on neonatal rat pups from P7 to P9 especially from an epigenetic perspective, the present study used the Morris water maze to detect cognitive deficits in spatial learning and memory, Sequenom methylation on the CpG island located in the promoter region of epidermal growth factor-containing fibulin-like extracellular matrix protein 1 (EFEMP1) to assess the methylation level of this region, and Western blot to measure the expression of EFEMP1, TIMP-3, and MMP-9. As the results have shown, repeated propofol administration on neonatal rats caused significant systemic growth retardation, impairment of spatial learning and memory, and hypermethylation of the CpG sites in the promoter region of EFEMP1 accompanied by lower expression of EFEMP1 and TIMP-3 and enhanced expression of MMP-9. These data suggest that repeated propofol administration in neonatal rats may generate hypermethylation in the promoter region of EFEMP1 which results in downregulation of the expression of EFEMP1 and tissue inhibitor of metalloproteinase-3 (TIMP-3) but upregulation of the expression of matrix metalloproteinase-9 (MMP-9), which together may affect the stability of ECM to hamper the development of the central nervous system and therefore lead to deficits in cognitive functions.
Collapse
|
26
|
Colàs-Campàs L, Farre J, Mauri-Capdevila G, Molina-Seguín J, Aymerich N, Ois Á, Roquer J, Tur S, García-Carreira MDC, Martí-Fàbregas J, Cruz-Culebras A, Segura T, Arque G, Purroy F. Inflammatory Response of Ischemic Tolerance in Circulating Plasma: Preconditioning-Induced by Transient Ischemic Attack (TIA) Phenomena in Acute Ischemia Patients (AIS). Front Neurol 2020; 11:552470. [PMID: 33192985 PMCID: PMC7658473 DOI: 10.3389/fneur.2020.552470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 09/30/2020] [Indexed: 11/15/2022] Open
Abstract
Introduction: Ischemic tolerance (IT) refers to a state where cells are resistant to the damaging effects caused by periods of ischemia. In a clinical scenario, the IT phenomenon would be activated by a recent transient ischemic attack (TIA) before an ischemic stroke (IS). The characterization of inflammatory protein expression patterns will contribute to improved understanding of IT. Methods: A total of 477 IS patients from nine hospitals, recruited between January 2011 and January 2016, were included in the current study and divided in three groups: 438 (91.9%) patients without previous TIA (group 1), 22 (4.6%) patients who suffered TIA 24 h before IS (group 2), and 17 (3.5%) patients who suffered TIA between 24 h and 7 days prior to IS (group 3). An inflammatory biomarker panel (IL-6, NT-proBNP, hsCRP, hs-Troponin, NSE, and S-100b) on plasma and a cytokine antibody array was performed to achieve the preconditioning signature potentially induced by TIA phenomena. Primary outcome was modified rankin scale (mRs) score at 90 days. Results: Recent previous TIA was associated with better clinical outcome at 90 days (median mRS of group 1: 2.0 [1.0–4.0]; group 2: 2.0 [0.0–3.0]; group 3: 1.0 [0–2.5]; p = 0.086) and smaller brain lesion (group 1: 3.7 [0.7–18.3]; group 2: 0.8 [0.3–8.9]; group 3: 0.6 [0.1–5.5] mL; p = 0.006). All inflammation biomarkers were down regulated in the groups of recent TIA prior to IS compared to those who did not suffer a TIA events. Moreover, a cytokine antibody array revealed 30 differentially expressed proteins between the three groups. Among them, HRG1-alpha (Fold change 74.4 between group 1 and 2; 74.2 between group 1 and 3) and MAC-1 (Fold change 0.05 between group 1 and 2; 0.06 between group 1 and 3) expression levels would better stratify patients with TIA 7 days before IS. These two proteins showed an earlier inflammation profile that was not detectable by the biomarker panel. Conclusion: Inflammatory pathways were activated by transient ischemic attack, however the period of time between this event and a further ischemic stroke could be determined by a protein signature that would contribute to define the role of ischemic tolerance induced by TIA.
Collapse
Affiliation(s)
- Laura Colàs-Campàs
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Joan Farre
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Medical Laboratory, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Gerard Mauri-Capdevila
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Jessica Molina-Seguín
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | | | | | | | - Silvia Tur
- Hospital Son Espases, Palma de Mallorca, Spain
| | | | | | | | - Tomás Segura
- Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Gloria Arque
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Francisco Purroy
- Clinical Neurosciences Group, Institut de Recerca Biomèdica de Lleida, Lleida, Spain.,Stroke Unit, Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| |
Collapse
|
27
|
Serifova X, Ugarte-Berzal E, Opdenakker G, Vandooren J. Homotrimeric MMP-9 is an active hitchhiker on alpha-2-macroglobulin partially escaping protease inhibition and internalization through LRP-1. Cell Mol Life Sci 2020; 77:3013-3026. [PMID: 31642940 PMCID: PMC11104829 DOI: 10.1007/s00018-019-03338-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/27/2019] [Accepted: 10/03/2019] [Indexed: 01/07/2023]
Abstract
Proteolysis is a crucial process in life, tightly controlled by numerous natural protease inhibitors. In human blood, alpha-2-macroglobulin is an emergency protease inhibitor preventing coagulation and damage to endothelia and leukocytes. With the use of a unique protease trapping mechanism, alpha-2-macroglobulin lures active proteases into its snap-trap, shields these from potential substrates and 'flags' their complex for elimination by receptor-mediated endocytosis. Matrix metalloprotease-9/gelatinase B is a secreted protease increased in blood of patients with inflammations, vascular disorders and cancers. Matrix metalloprotease-9 occurs as monomers and stable homotrimers, but the reason for their co-existence remains obscure. We discovered that matrix metalloprotease-9 homotrimers undergo reduced anti-proteolytic regulation by alpha-2-macroglobulin and are able to travel as a proteolytically active hitchhiker on alpha-2-macroglobulin. As a comparison, we revealed that monomeric active matrix metalloprotease-9 is efficiently trapped by human plasma alpha-2-macroglobulin and this masks the detection of activated matrix metalloprotease-9 with standard analysis techniques. In addition, we show that alpha-2-macroglobulin/trimer complexes escape clearance through the receptor low-density lipoprotein receptor-related protein 1, also known as the alpha-2-macroglobulin receptor. Thus, the biochemistry and biology of matrix metalloprotease-9 monomers and trimers are completely different as multimerization enables active matrix metalloprotease-9 to partially avoid alpha-2-macroglobulin regulation both by direct protease inhibition and by removal from the extracellular space by receptor-mediated endocytosis. Finally, for the biomarker field, the analysis of alpha-2-macroglobulin/protease complexes with upgraded technology is advocated as a quotum for protease activation in human plasma samples.
Collapse
Affiliation(s)
- Xena Serifova
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Herestraat 49, Bus 1044, 3000, Leuven, Belgium
| | - Estefania Ugarte-Berzal
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Herestraat 49, Bus 1044, 3000, Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Herestraat 49, Bus 1044, 3000, Leuven, Belgium
| | - Jennifer Vandooren
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, KU Leuven, Herestraat 49, Bus 1044, 3000, Leuven, Belgium.
| |
Collapse
|
28
|
Amruta N, Rahman AA, Pinteaux E, Bix G. Neuroinflammation and fibrosis in stroke: The good, the bad and the ugly. J Neuroimmunol 2020; 346:577318. [PMID: 32682140 PMCID: PMC7794086 DOI: 10.1016/j.jneuroim.2020.577318] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023]
Abstract
Stroke is the leading cause of death and the main cause of disability in surviving patients. The detrimental interaction between immune cells, glial cells, and matrix components in stroke pathology results in persistent inflammation that progresses to fibrosis. A substantial effort is being directed toward understanding the exact neuroinflammatory events that take place as a result of stroke. The initiation of a potent cytokine response, along with immune cell activation and infiltration in the ischemic core, has massive acute deleterious effects, generally exacerbated by comorbid inflammatory conditions. There is secondary neuroinflammation that promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. This highlights the need for a better understanding of the neuroinflammatory and fibrotic processes, as well as the need to identify new mechanisms and potential modulators. In this review, we summarize several aspects of stroke-induced inflammation, fibrosis, and include a discussion of cytokine inhibitors/inducers, immune cells, and fibro-inflammation signaling inhibitors in order to identify new pharmacological means of intervention.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Abir A Rahman
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
29
|
Silva NJ, Nagashima M, Li J, Kakuk‐Atkins L, Ashrafzadeh M, Hyde DR, Hitchcock PF. Inflammation and matrix metalloproteinase 9 (Mmp-9) regulate photoreceptor regeneration in adult zebrafish. Glia 2020; 68:1445-1465. [PMID: 32034934 PMCID: PMC7317489 DOI: 10.1002/glia.23792] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/24/2019] [Accepted: 01/23/2020] [Indexed: 12/17/2022]
Abstract
Brain injury activates complex inflammatory signals in dying neurons, surviving neurons, and glia. Here, we establish that inflammation regulates the regeneration of photoreceptors in the zebrafish retina and determine the cellular expression and function of the inflammatory protease, matrix metalloproteinase 9 (Mmp-9), during this regenerative neurogenesis. Following photoreceptor ablation, anti-inflammatory treatment suppresses the number of injury-induced progenitors and regenerated photoreceptors. Upon photoreceptor injury, mmp-9 is induced in Müller glia and Müller glia-derived photoreceptor progenitors. Deleting mmp-9 results in over production of injury-induced progenitors and regenerated photoreceptors, but over time the absence of Mmp-9 compromises the survival of the regenerated cones. At all time-points studied, the levels of tnf-α are significantly elevated in mutant retinas. Anti-inflammatory treatment in mutants rescues the defects in cone survival. These data provide a link between injury-induced inflammation in the vertebrate CNS, Mmp-9 function during neuronal regeneration and the requirement of Mmp-9 for the survival of regenerated cones.
Collapse
Affiliation(s)
- Nicholas J. Silva
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMichigan
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - Mikiko Nagashima
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - Jingling Li
- Department of Biological SciencesUniversity of Notre DameNotre DameIndiana
| | - Laura Kakuk‐Atkins
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - Milad Ashrafzadeh
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| | - David R. Hyde
- Department of Biological SciencesUniversity of Notre DameNotre DameIndiana
| | - Peter F. Hitchcock
- Neuroscience Graduate ProgramUniversity of MichiganAnn ArborMichigan
- Department of Ophthalmology and Visual SciencesUniversity of MichiganAnn ArborMichigan
| |
Collapse
|
30
|
Electrospun Resveratrol-Loaded Polyvinylpyrrolidone/Cyclodextrin Nanofibers and Their Biomedical Applications. Pharmaceutics 2020; 12:pharmaceutics12060552. [PMID: 32545836 PMCID: PMC7357065 DOI: 10.3390/pharmaceutics12060552] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Resveratrol is a naturally occurring polyphenol compound which has been shown to possess antioxidant and anti-inflammatory properties. However, its pharmaceutical applications are limited by its poor water solubility. In this study, we used electrospinning technology to synthesize nanofibers of polyvinylpyrrolidone (PVP) and hydroxypropyl-β-cyclodextrin (HPBCD) loaded with resveratrol. We used X-ray diffractometry to analyze crystalline structure, Fourier transform infrared spectroscopy to determine intermolecular hydrogen bonding, antioxidant assays to measure antioxidant activity, and Franz diffusion cells to evaluate skin penetration. Our results showed that the aqueous solubility of resveratrol nanofibers was greatly improved (by more than 20,000-fold) compared to the pure compound. Analysis of physicochemical properties demonstrated that following nanofiber formation, resveratrol was converted from a crystalline to amorphous structure, and resveratrol formed new intermolecular bonds with PVP and HPBCD. Moreover, resveratrol nanofibers showed good antioxidant activity. In addition, the skin penetration ability of resveratrol in the nanofiber formulation was greater than that of pure resveratrol. Furthermore, resveratrol nanofibers suppressed particulate matter (PM)-induced expression of inflammatory proteins (COX-2 and MMP-9) in HaCaT keratinocytes. Therefore, resveratrol-loaded nanofibers can effectively improve the solubility and physicochemical properties of resveratrol, and may have potential applications as an antioxidant and anti-inflammatory formulation for topical skin application.
Collapse
|
31
|
Dao T, Salahuddin S, Charfi C, Sicard AA, Jenabian MA, Annabi B. Pharmacological targeting of neurotensin response by diet-derived EGCG in macrophage-differentiated HL-60 promyelocytic leukemia cells. PHARMANUTRITION 2020; 12:100191. [DOI: 10.1016/j.phanu.2020.100191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Doxycycline improves traumatic brain injury outcomes in a murine survival model. J Trauma Acute Care Surg 2020; 89:435-440. [DOI: 10.1097/ta.0000000000002801] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
33
|
Chen S, Chen H, Du Q, Shen J. Targeting Myeloperoxidase (MPO) Mediated Oxidative Stress and Inflammation for Reducing Brain Ischemia Injury: Potential Application of Natural Compounds. Front Physiol 2020; 11:433. [PMID: 32508671 PMCID: PMC7248223 DOI: 10.3389/fphys.2020.00433] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress and inflammation are two critical pathological processes of cerebral ischemia-reperfusion injury. Myeloperoxidase (MPO) is a critical inflammatory enzyme and therapeutic target triggering both oxidative stress and neuroinflammation in the pathological process of cerebral ischemia-reperfusion injury. MPO is presented in infiltrated neutrophils, activated microglial cells, neurons, and astrocytes in the ischemic brain. Activation of MPO can catalyze the reaction of chloride and H2O2 to produce HOCl. MPO also mediates oxidative stress by promoting the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), modulating the polarization and inflammation-related signaling pathways in microglia and neutrophils. MPO can be a therapeutic target for attenuating oxidative damage and neuroinflammation in ischemic stroke. Targeting MPO with inhibitors or gene deficiency significantly reduced brain infarction and improved neurological outcomes. This article discusses the important roles of MPO in mediating oxidative stress and neuroinflammation during cerebral ischemia-reperfusion injury and reviews the current understanding of the underlying mechanisms. Furthermore, we summarize the active compounds from medicinal herbs with potential as MPO inhibitors for anti-oxidative stress and anti-inflammation to attenuate cerebral ischemia-reperfusion injury, and as adjunct therapeutic agents for extending the window of thrombolytic treatment. We highlight that targeting MPO could be a promising strategy for alleviating ischemic brain injury, which merits further translational study.
Collapse
Affiliation(s)
- Shuang Chen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Qiaohui Du
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| |
Collapse
|
34
|
Chen H, He Y, Chen S, Qi S, Shen J. Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: Applications for natural product efficacy with omics and systemic biology. Pharmacol Res 2020; 158:104877. [PMID: 32407958 DOI: 10.1016/j.phrs.2020.104877] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Oxidative/nitrosative stress and neuroinflammation are critical pathological processes in cerebral ischemia-reperfusion injury, and their intimate interactions mediate neuronal damage, blood-brain barrier (BBB) damage and hemorrhagic transformation (HT) during ischemic stroke. We review current progress towards understanding the interactions of oxidative/nitrosative stress and inflammatory responses in ischemic brain injury. The interactions between reactive oxygen species (ROS)/reactive nitrogen species (RNS) and innate immune receptors such as TLR2/4, NOD-like receptor, RAGE, and scavenger receptors are crucial pathological mechanisms that amplify brain damage during cerebral ischemic injury. Furthermore, we review the current progress of omics and systematic biology approaches for studying complex network regulations related to oxidative/nitrosative stress and inflammation in the pathology of ischemic stroke. Targeting oxidative/nitrosative stress and neuroinflammation could be a promising therapeutic strategy for ischemic stroke treatment. We then review recent advances in discovering compounds from medicinal herbs with the bioactivities of simultaneously regulating oxidative/nitrosative stress and pro-inflammatory molecules for minimizing ischemic brain injury. These compounds include sesamin, baicalin, salvianolic acid A, 6-paradol, silymarin, apocynin, 3H-1,2-Dithiole-3-thione, (-)-epicatechin, rutin, Dl-3-N-butylphthalide, and naringin. We finally summarize recent developments of the omics and systematic biology approaches for exploring the molecular mechanisms and active compounds of Traditional Chinese Medicine (TCM) formulae with the properties of antioxidant and anti-inflammation for neuroprotection. The comprehensive omics and systematic biology approaches provide powerful tools for exploring therapeutic principles of TCM formulae and developing precision medicine for stroke treatment.
Collapse
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Yacong He
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Shuang Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China; School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
35
|
Buchheit T, Huh Y, Maixner W, Cheng J, Ji RR. Neuroimmune modulation of pain and regenerative pain medicine. J Clin Invest 2020; 130:2164-2176. [PMID: 32250346 PMCID: PMC7190995 DOI: 10.1172/jci134439] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Regenerative pain medicine, which seeks to harness the body's own reparative capacity, is rapidly emerging as a field within pain medicine and orthopedics. It is increasingly appreciated that common analgesic mechanisms for these treatments depend on neuroimmune modulation. In this Review, we discuss recent progress in mechanistic understanding of nociceptive sensitization in chronic pain with a focus on neuroimmune modulation. We also examine the spectrum of regenerative outcomes, including preclinical and clinical outcomes. We further distinguish the analgesic mechanisms of regenerative therapies from those of cellular replacement, creating a conceptual and mechanistic framework to evaluate future research on regenerative medicine.
Collapse
Affiliation(s)
- Thomas Buchheit
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Anesthesiology Service, Durham Veterans Affairs Health Care System, Durham, North Carolina, USA
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - William Maixner
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jianguo Cheng
- Departments of Pain Management and Neurosciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
36
|
Hamanaka G, Kubo T, Ohtomo R, Takase H, Reyes-Bricio E, Oribe S, Osumi N, Lok J, Lo EH, Arai K. Microglial responses after phagocytosis: Escherichia coli bioparticles, but not cell debris or amyloid beta, induce matrix metalloproteinase-9 secretion in cultured rat primary microglial cells. Glia 2020; 68:1435-1444. [PMID: 32057146 DOI: 10.1002/glia.23791] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 12/13/2022]
Abstract
Upon infection or brain damage, microglia are activated to play roles in immune responses, including phagocytosis and soluble factor release. However, little is known whether the event of phagocytosis could be a trigger for releasing soluble factors from microglia. In this study, we tested if microglia secrete a neurovascular mediator matrix metalloproteinase-9 (MMP-9) after phagocytosis in vitro. Primary microglial cultures were prepared from neonatal rat brains. Cultured microglia phagocytosed Escherichia coli bioparticles within 2 hr after incubation and started to secrete MMP-9 at around 12 hr after the phagocytosis. A TLR4 inhibitor TAK242 suppressed the E. coli-bioparticle-induced MMP-9 secretion. However, TAK242 did not change the engulfment of E. coli bioparticles in microglial cultures. Because lipopolysaccharides (LPS), the major component of the outer membrane of E. coli, also induced MMP-9 secretion in a dose-response manner and because the response was inhibited by TAK242 treatment, we assumed that the LPS-TLR4 pathway, which was activated by adhering to the substance, but not through the engulfing process of phagocytosis, would play a role in releasing MMP-9 from microglia after E. coli bioparticle treatment. To support the finding that the engulfing step would not be a critical trigger for MMP-9 secretion after the event of phagocytosis in microglia, we confirmed that cell debris and amyloid beta were both captured into microglia via phagocytosis, but neither of them induced MMP-9 secretion from microglia. Taken together, these data demonstrate that microglial response in MMP-9 secretion after phagocytosis differs depending on the types of particles/substances that microglia encountered.
Collapse
Affiliation(s)
- Gen Hamanaka
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tomoya Kubo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ryo Ohtomo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hajime Takase
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Estefania Reyes-Bricio
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shuntaro Oribe
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, United Centers for Advanced Research and Translational Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Josephine Lok
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts
| | - Eng H Lo
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ken Arai
- Neuroprotection Research Laboratory, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.,Neuroprotection Research Laboratory, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
37
|
Hardy E, Fernandez-Patron C. Destroy to Rebuild: The Connection Between Bone Tissue Remodeling and Matrix Metalloproteinases. Front Physiol 2020; 11:47. [PMID: 32116759 PMCID: PMC7013034 DOI: 10.3389/fphys.2020.00047] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Bone is a dynamic organ that undergoes constant remodeling, an energetically costly process by which old bone is replaced and localized bone defects are repaired to renew the skeleton over time, thereby maintaining skeletal health. This review provides a general overview of bone’s main players (bone lining cells, osteocytes, osteoclasts, reversal cells, and osteoblasts) that participate in bone remodeling. Placing emphasis on the family of extracellular matrix metalloproteinases (MMPs), we describe how: (i) Convergence of multiple protease families (including MMPs and cysteine proteinases) ensures complexity and robustness of the bone remodeling process, (ii) Enzymatic activity of MMPs affects bone physiology at the molecular and cellular levels and (iii) Either overexpression or deficiency/insufficiency of individual MMPs impairs healthy bone remodeling and systemic metabolism. Today, it is generally accepted that proteolytic activity is required for the degradation of bone tissue in osteoarthritis and osteoporosis. However, it is increasingly evident that inactivating mutations in MMP genes can also lead to bone pathology including osteolysis and metabolic abnormalities such as delayed growth. We argue that there remains a need to rethink the role played by proteases in bone physiology and pathology.
Collapse
Affiliation(s)
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
38
|
Abstract
BACKGROUND The integrity of the blood-brain barrier (BBB) is paramount in limiting vasogenic edema following traumatic brain injury (TBI). The purpose of this study was to ascertain if quetiapine, an atypical antipsychotic commonly used in trauma/critical care for delirium, protects the BBB and attenuates hyperpermeability in TBI. METHODS The effect of quetiapine on hyperpermeability was examined through molecular modeling, cellular models in vitro and small animal models in vivo. Molecular docking was performed with AutoDock Vina to matrix metalloproteinase-9. Rat brain microvascular endothelial cells (BMECs) were pretreated with quetiapine (20 μM; 1 hour) followed by an inflammatory activator (20 μg/mL chitosan; 2 hours) and compared to controls. Immunofluorescence localization for tight junction proteins zonula occludens-1 and adherens junction protein β-catenin was performed. Human BMECs were grown as a monolayer and pretreated with quetiapine (20 μM; 1 hour) followed by chitosan (20 μg/mL; 2 hours), and transendothelial electrical resistance was measured. C57BL/6 mice (n = 5/group) underwent mild to moderate TBI (controlled cortical impactor) or sham craniotomy. The treatment group was given 10 mg/kg quetiapine intravenously 10 minutes after TBI. The difference in fluorescence intensity between intravascular and interstitium (ΔI) represented BBB hyperpermeability. A matrix metalloproteinase-9 activity assay was performed in brain tissue from animals in the experimental groups ex vivo. RESULTS In silico studies showed quetiapine thermodynamically favorable binding to MMP-9. Junctional localization of zonula occludens-1 and β-catenin showed retained integrity in quetiapine-treated cells as compared with the chitosan group in rat BMECs. Quetiapine attenuated monolayer permeability compared with chitosan group (p < 0.05) in human BMECs. In the animal studies, there was a significant decrease in BBB hyperpermeability and MMP-9 activity when compared between the TBI and TBI plus quetiapine groups (p < 0.05). CONCLUSION Quetiapine treatment may have novel anti-inflammatory properties to provide protection to the BBB by preserving tight junction integrity. LEVEL OF EVIDENCE level IV.
Collapse
|
39
|
Michalovicz LT, Kelly KA, Vashishtha S, Ben‐Hamo R, Efroni S, Miller JV, Locker AR, Sullivan K, Broderick G, Miller DB, O’Callaghan JP. Astrocyte-specific transcriptome analysis using the ALDH1L1 bacTRAP mouse reveals novel biomarkers of astrogliosis in response to neurotoxicity. J Neurochem 2019; 150:420-440. [PMID: 31222732 PMCID: PMC6771645 DOI: 10.1111/jnc.14800] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 12/21/2022]
Abstract
Neurotoxicology is hampered by the inability to predict regional and cellular targets of toxicant-induced damage. Evaluating astrogliosis overcomes this problem because reactive astrocytes highlight the location of toxicant-induced damage. While enhanced expression of glial fibrillary acidic protein is a hallmark of astrogliosis, few other biomarkers have been identified. However, bacterial artificial chromosome - translating ribosome affinity purification (bacTRAP) technology allows for characterization of the actively translating transcriptome of a particular cell type; use of this technology in aldehyde dehydrogenase 1 family member L1 (ALDH1L1) bacTRAP mice can identify genes selectively expressed in astrocytes. The aim of this study was to characterize additional biomarkers of neurotoxicity-induced astrogliosis using ALDH1L1 bacTRAP mice. The known dopaminergic neurotoxicant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 12.5 mg/kg s.c.) was used to induce astrogliosis. Striatal tissue was obtained 12, 24, and 48 h following exposure for the isolation of actively translating RNA. Subsequently, MPTP-induced changes in this RNA pool were analyzed by microarray and 184 statistically significant, differentially expressed genes were identified. The dataset was interrogated by gene ontology, pathway, and co-expression network analyses, which identified novel genes, as well as those with known immune and inflammatory functions. Using these analyses, we were directed to several genes associated with reactive astrocytes. Of these, TIMP1 and miR-147 were identified as candidate biomarkers because of their robust increased expression following both MPTP and trimethyl tin exposures. Thus, we have demonstrated that bacTRAP can be used to identify new biomarkers of astrogliosis and aid in the characterization of astrocyte phenotypes induced by toxicant exposures. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/. Cover Image for this issue: doi: 10.1111/jnc.14518.
Collapse
Affiliation(s)
- Lindsay T. Michalovicz
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - Kimberly A. Kelly
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - Saurabh Vashishtha
- Center for Clinical Systems BiologyRochester General Hospital Research InstituteRochesterNew YorkUSA
| | - Rotem Ben‐Hamo
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Sol Efroni
- The Mina and Everard Goodman Faculty of Life SciencesBar‐Ilan UniversityRamat‐GanIsrael
| | - Julie V. Miller
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - Alicia R. Locker
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | | | - Gordon Broderick
- Center for Clinical Systems BiologyRochester General Hospital Research InstituteRochesterNew YorkUSA
| | - Diane B. Miller
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| | - James P. O’Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and PreventionNational Institute for Occupational Safety and HealthMorgantownWest VirginiaUSA
| |
Collapse
|
40
|
Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation 2019; 16:142. [PMID: 31291966 PMCID: PMC6617684 DOI: 10.1186/s12974-019-1516-2] [Citation(s) in RCA: 892] [Impact Index Per Article: 148.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Stroke, the third leading cause of death and disability worldwide, is undergoing a change in perspective with the emergence of new ideas on neurodegeneration. The concept that stroke is a disorder solely of blood vessels has been expanded to include the effects of a detrimental interaction between glia, neurons, vascular cells, and matrix components, which is collectively referred to as the neurovascular unit. Following the acute stroke, the majority of which are ischemic, there is secondary neuroinflammation that both promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. The proinflammatory signals from immune mediators rapidly activate resident cells and influence infiltration of a wide range of inflammatory cells (neutrophils, monocytes/macrophages, different subtypes of T cells, and other inflammatory cells) into the ischemic region exacerbating brain damage. In this review, we discuss how neuroinflammation has both beneficial as well as detrimental roles and recent therapeutic strategies to combat pathological responses. Here, we also focus on time-dependent entry of immune cells to the ischemic area and the impact of other pathological mediators, including oxidative stress, excitotoxicity, matrix metalloproteinases (MMPs), high-mobility group box 1 (HMGB1), arachidonic acid metabolites, mitogen-activated protein kinase (MAPK), and post-translational modifications that could potentially perpetuate ischemic brain damage after the acute injury. Understanding the time-dependent role of inflammatory factors could help in developing new diagnostic, prognostic, and therapeutic neuroprotective strategies for post-stroke inflammation.
Collapse
Affiliation(s)
- Richard L. Jayaraj
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Sheikh Azimullah
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Fakhreya Y. Jalal
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Gary A. Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131 USA
| |
Collapse
|
41
|
MMP-9 Contributes to Dendritic Spine Remodeling Following Traumatic Brain Injury. Neural Plast 2019; 2019:3259295. [PMID: 31198417 PMCID: PMC6526556 DOI: 10.1155/2019/3259295] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 04/03/2019] [Indexed: 01/30/2023] Open
Abstract
Traumatic brain injury (TBI) occurs when a blow to the head causes brain damage. Apart from physical trauma, it causes a wide range of cognitive, behavioral, and emotional deficits including impairments in learning and memory. On neuronal level, TBI may lead to circuitry remodeling and in effect imbalance between excitatory and inhibitory neurotransmissions. Such change in brain homeostasis may often lead to brain disorders. The basic units of neuronal connectivity are dendritic spines that are tiny protrusions forming synapses between two cells in a network. Spines are dynamic structures that undergo morphological transformation throughout life. Their shape is strictly related to an on/off state of synapse and the strength of synaptic transmission. Matrix metalloproteinase-9 (MMP-9) is an extrasynaptically operating enzyme that plays a role in spine remodeling and has been reported to be activated upon TBI. The aim of the present study was to evaluate the influence of MMP-9 on dendritic spine density and morphology following controlled cortical impact (CCI) as animal model of TBI. We examined spine density and dendritic spine shape in the cerebral cortex and the hippocampus. CCI caused a marked decrease in spine density as well as spine shrinkage in the cerebral cortex ipsilateral to the injury, when compared to sham animals and contralateral side both 1 day and 1 week after the insult. Decreased spine density was also observed in the dentate gyrus of the hippocampus; however, in contrast to the cerebral cortex, spines in the DG became more filopodia-like. In mice lacking MMP-9, no effects of TBI on spine density and morphology were observed.
Collapse
|
42
|
Gao N, Guo T, Luo H, Tu G, Niu F, Yan M, Xia Y. Association of the MMP-9 polymorphism and ischemic stroke risk in southern Chinese Han population. BMC Neurol 2019; 19:67. [PMID: 30992065 PMCID: PMC6469199 DOI: 10.1186/s12883-019-1285-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 03/26/2019] [Indexed: 02/05/2023] Open
Abstract
Background Stroke is a serious cardiovascular disease and is also the leading cause of long-term disability in developing and developed countries. Because matrix metalloproteinase-9 (MMP-9) is associated with the risk of many cardiovascular diseases, we investigated the relationship between single nucleotide polymorphisms (SNPs) in MMP-9 and the risk of Ischemic stroke (IS) in a southern Chinese Han population. Methods This study included 250 stroke patients and 250 healthy controls. Genotyping was performed using the Agena MassARRAY system, and chi-squared tests and genetic models were used to evaluate the associations between MMP-9 SNPs and the risk of IS. Odds ratio (OR) and 95% confidence intervals (CIs) were calculated by unconditional logistic regression adjusted for age. Results Polymorphism rs3787268 was associated with increased the risk of IS. Specifically, the genotype “G/A” significantly correlated with IS risk in the co-dominant model [odds ratio (OR) = 1.62; 95% confidence interval (CI) = 1.10–2.41; p = 0.035)], while genotypes “G/A” and “A/A” may increase the risk of IS based on the dominant model (OR = 1.62; 95% CI = 1.12–2.35; p = 0.0097). This SNP was also significantly associated with IS risk in the log-additive model (OR = 1.33; 95% CI = 1.03–1.70; p = 0.026). Conversely, haplotype “C/G” appears to reduce the risk of IS (OR = 0.71; 95% CI = 0.54–0.95; p = 0.019). Conclusions Our study showed that the rs3787268 locus in the MMP-9 gene may increase risk of IS in a southern Chinese Han population and thus provide insight into the IS pathogenesis.
Collapse
Affiliation(s)
- Ning Gao
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical College in Central South University, Haikou, Hainan, 570208, China
| | - Tie Guo
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Han Luo
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical College in Central South University, Haikou, Hainan, 570208, China
| | - Guolong Tu
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical College in Central South University, Haikou, Hainan, 570208, China
| | - Fanglin Niu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, Shaanxi, China
| | - Mengdan Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Northwest University, Ministry of Education, Xi'an, Shaanxi, China
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital of Xiangya Medical College in Central South University, Haikou, Hainan, 570208, China.
| |
Collapse
|
43
|
Gao F, Zhang Q, Li Y, Tai Y, Xin X, Wang X, Wang Q. Transcutaneous electrical acupoint stimulation for prevention of postoperative delirium in geriatric patients with silent lacunar infarction: a preliminary study. Clin Interv Aging 2018; 13:2127-2134. [PMID: 30425466 PMCID: PMC6205526 DOI: 10.2147/cia.s183698] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose This study aims to investigate the effect of transcutaneous electrical acupoint stimulation (TEAS) on postoperative delirium (POD) in elderly patients with silent lacunar infarct and preliminarily to determine the relationship among TEAS, blood–brain barrier (BBB), neuroinflammation, and POD. Patients and methods Sixty-four-old patients with silent lacunar infarct were randomly divided into two groups: group TEAS and control group (group C). Patients in the group TEAS received TEAS (disperse-dense waves; frequency, 2/100 Hz) on acupoints Hegu and Neiguan of both sides starting from 30 minutes before induction of anesthesia until the end of surgery, and the intensity was the maximum current that could be tolerated. In group C, electrodes were placed on the same acupoints before anesthesia induction, but no current was given. At 0 minute before the treatment of TEAS, 30 minutes after skin incision, and after completion of surgery (T1–3), blood samples were extracted to detect the concentration of serum tumor necrosis factor (TNF)-α, interleukin-6 (IL-6), matrix metalloproteinase-9 (MMP-9), and S100β. We assessed patients for delirium and coma twice daily in the first 3 postoperative days using the Confusion Assessment Method for the intensive care unit and the Richmond Agitation-Sedation Scale. Results This study preliminarily suggests that TEAS can reduce the development of POD in elderly patients with silent lacunar infarction (6.3% vs 25.0%; P=0.039). Compared with the baseline value at T1, the serum concentrations of IL-6, TNF-α, MMP-9, and S100β were significantly increased at T2–3 in both the groups (P<0.05). Compared with group TEAS, serum levels of TNF-α and IL-6 were higher at T2–3 and serum levels of MMP-9 and S100β were higher at T3 in group C (P<0.05). The intraoperative anesthetic consumptions were less in group TEAS than group C. Conclusion TEAS can alleviate POD in older patients with silent lacunar infarction and may be related to reduce the neuroinflammation by lowering the permeability of BBB.
Collapse
Affiliation(s)
- Fang Gao
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China,
| | - Qi Zhang
- Department of Anesthesiology, Children's Hospital of Hebei Province, Shijiazhuang City, Hebei, People's Republic of China
| | - Yanan Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China,
| | - Yanlei Tai
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China,
| | - Xi Xin
- Department of Anesthesiology, Tianjin Third Central Hospital, Tianjin, People's Republic of China
| | - Xiuli Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China,
| | - Qiujun Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang City, Hebei, People's Republic of China,
| |
Collapse
|
44
|
Guvercin G, Karakus V, Aksit M, Dere Y, Aktar M, Alpay H, Bozkaya G, Tatar E. Matrix metalloproteinase-9, 10, and stress hyperglycaemia in acute kidney injury. Eur J Clin Invest 2018; 48:e12963. [PMID: 29856477 DOI: 10.1111/eci.12963] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 05/30/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND This study investigated the effect of matrix metalloproteinase (MMP)-9 and 10, and stress hyperglycaemia on the necessity of emergency renal replacement therapy (RRT) and mortality in nondiabetic geriatric patients with acute kidney injury (AKI). MATERIALS AND METHODS The present observational and longitudinal study included 101 nondiabetic geriatric patients (age >65 years) with AKI. The serum levels of MMP-9 and MMP-10 were evaluated in these patients. Serum glucose level >140 mg/dL at the time of admission was accepted as stress hyperglycaemia. RESULTS The average age of patients was 81 ± 7.1 years. Stress hyperglycaemia was diagnosed in 34.6% of the cases; the majority of these cases were patients with high-serum urea, CRP, and chronic kidney disease. The average levels of MMP-9 and MMP-10 were found to be 199 ± 38 and 16.5 ± 7.5 ng/mL, respectively. Thirty-one cases (30.6%) mortality during hospitalization and 20 cases (20%) underwent emergency RRT. Multiregression analysis showed the serum urea (P < .001) and stress hyperglycaemia (P = .03) to be independently associated with mortality. Also, serum urea (P = .01), potassium level (P = .03), and MMP-10 levels (P = .03) were independently associated with the necessity of the emergency RRT. The MMP-9 levels exhibited no relation with the necessity of emergency RRT and mortality. CONCLUSION Stress hyperglycaemia is a common condition among nondiabetic geriatric patients with AKI and is related to mortality. Serum MMP-10 levels serve as an important predictor of the necessity of emergency RRT in these patients.
Collapse
Affiliation(s)
- Guray Guvercin
- Department of Internal Medicine, Izmir Bozyaka Education and Research Hospital, Health Sciences University, Izmir, Turkey
| | - Volkan Karakus
- Division of Hematology, Mugla Sitki Kocman University Training and Research Hospital, Mugla, Turkey
| | - Murat Aksit
- Department of Biochemistry, Izmir Bozyaka Education and Research Hospital, Health Sciences University, Izmir, Turkey
| | - Yelda Dere
- Division of Pathology, Mugla Sitki Kocman University Training and Research Hospital, Mugla, Turkey
| | - Merve Aktar
- Department of Internal Medicine, Izmir Bozyaka Education and Research Hospital, Health Sciences University, Izmir, Turkey
| | - Hasan Alpay
- Department of Internal Medicine, Izmir Bozyaka Education and Research Hospital, Health Sciences University, Izmir, Turkey
| | - Giray Bozkaya
- Department of Biochemistry, Izmir Bozyaka Education and Research Hospital, Health Sciences University, Izmir, Turkey
| | - Erhan Tatar
- Department of Nephrology, Izmir Bozyaka Education and Research Hospital, Health Sciences University, Izmir, Turkey
| |
Collapse
|
45
|
Cheng Z, Wang L, Qu M, Liang H, Li W, Li Y, Deng L, Zhang Z, Yang GY. Mesenchymal stem cells attenuate blood-brain barrier leakage after cerebral ischemia in mice. J Neuroinflammation 2018; 15:135. [PMID: 29724240 PMCID: PMC5932816 DOI: 10.1186/s12974-018-1153-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
Background Ischemic stroke induced matrixmetallo-proteinase-9 (MMP-9) upregulation, which increased blood-brain barrier permeability. Studies demonstrated that mesenchymal stem cell therapy protected blood-brain barrier disruption from several cerebrovascular diseases. However, the underlying mechanism was largely unknown. We therefore hypothesized that mesenchymal stem cells reduced blood-brain barrier destruction by inhibiting matrixmetallo-proteinase-9 and it was related to intercellular adhesion molecule-1 (ICAM-1). Methods Adult ICR male mice (n = 118) underwent 90-min middle cerebral artery occlusion and received 2 × 105 mesenchymal stem cell transplantation. Neurobehavioral outcome, infarct volume, and blood-brain barrier permeability were measured after ischemia. The relationship between myeloperoxidase (MPO) activity and ICAM-1 release was further determined. Results We found that intracranial injection of mesenchymal stem cells reduced infarct volume and improved behavioral function in experimental stroke models (p < 0.05). IgG leakage, tight junction protein loss, and inflammatory cytokines IL-1β, IL-6, and TNF-α reduced in mesenchymal stem cell-treated mice compared to the control group following ischemia (p < 0.05). After transplantation, MMP-9 was decreased in protein and activity levels as compared with controls (p < 0.05). Furthermore, myeloperoxidase-positive cells and myeloperoxidase activity were decreased in mesenchymal stem cell-treated mice (p < 0.05). Conclusion The results showed that mesenchymal stem cell therapy attenuated blood-brain barrier disruption in mice after ischemia. Mesenchymal stem cells attenuated the upward trend of MMP-9 and potentially via downregulating ICAM-1 in endothelial cells. Adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) pathway may influence MMP-9 expression of neutrophils and resident cells, and ICAM-1 acted as a key factor in the paracrine actions of mesenchymal stem cell.
Collapse
Affiliation(s)
- Zhuo Cheng
- School of Biomedical Engineering and Shanghai Jiao Tong University affiliated sixth people's hospital, Shanghai Jiao Tong University, Shanghai, 200000, China
| | - Liping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Meijie Qu
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Huaibin Liang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Wanlu Li
- School of Biomedical Engineering and Shanghai Jiao Tong University affiliated sixth people's hospital, Shanghai Jiao Tong University, Shanghai, 200000, China
| | - Yongfang Li
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Lidong Deng
- School of Biomedical Engineering and Shanghai Jiao Tong University affiliated sixth people's hospital, Shanghai Jiao Tong University, Shanghai, 200000, China
| | - Zhijun Zhang
- School of Biomedical Engineering and Shanghai Jiao Tong University affiliated sixth people's hospital, Shanghai Jiao Tong University, Shanghai, 200000, China
| | - Guo-Yuan Yang
- School of Biomedical Engineering and Shanghai Jiao Tong University affiliated sixth people's hospital, Shanghai Jiao Tong University, Shanghai, 200000, China. .,Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
46
|
Chen HS, Chen X, Li WT, Shen JG. Targeting RNS/caveolin-1/MMP signaling cascades to protect against cerebral ischemia-reperfusion injuries: potential application for drug discovery. Acta Pharmacol Sin 2018; 39:669-682. [PMID: 29595191 PMCID: PMC5943912 DOI: 10.1038/aps.2018.27] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/26/2018] [Indexed: 02/07/2023]
Abstract
Reactive nitrogen species (RNS) play important roles in mediating cerebral ischemia-reperfusion injury. RNS activate multiple signaling pathways and participate in different cellular events in cerebral ischemia-reperfusion injury. Recent studies have indicated that caveolin-1 and matrix metalloproteinase (MMP) are important signaling molecules in the pathological process of ischemic brain injury. During cerebral ischemia-reperfusion, the production of nitric oxide (NO) and peroxynitrite (ONOO−), two representative RNS, down-regulates the expression of caveolin-1 (Cav-1) and, in turn, further activates nitric oxide synthase (NOS) to promote RNS generation. The increased RNS further induce MMP activation and mediate disruption of the blood-brain barrier (BBB), aggravating the brain damage in cerebral ischemia-reperfusion injury. Therefore, the feedback interaction among RNS/Cav-1/MMPs provides an amplified mechanism for aggravating ischemic brain damage during cerebral ischemia-reperfusion injury. Targeting the RNS/Cav-1/MMP pathway could be a promising therapeutic strategy for protecting against cerebral ischemia-reperfusion injury. In this mini-review article, we highlight the important role of the RNS/Cav-1/MMP signaling cascades in ischemic stroke injury and review the current progress of studies seeking therapeutic compounds targeting the RNS/Cav-1/MMP signaling cascades to attenuate cerebral ischemia-reperfusion injury. Several representative natural compounds, including calycosin-7-O-β-D-glucoside, baicalin, Momordica charantia polysaccharide (MCP), chlorogenic acid, lutein and lycopene, have shown potential for targeting the RNS/Cav-1/MMP signaling pathway to protect the brain in ischemic stroke. Therefore, the RNS/Cav-1/MMP pathway is an important therapeutic target in ischemic stroke treatment.
Collapse
|
47
|
Brzdak P, Nowak D, Wiera G, Mozrzymas JW. Multifaceted Roles of Metzincins in CNS Physiology and Pathology: From Synaptic Plasticity and Cognition to Neurodegenerative Disorders. Front Cell Neurosci 2017; 11:178. [PMID: 28713245 PMCID: PMC5491558 DOI: 10.3389/fncel.2017.00178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) and membrane proteolysis play a key role in structural and functional synaptic plasticity associated with development and learning. A growing body of evidence underscores the multifaceted role of members of the metzincin superfamily, including metalloproteinases (MMPs), A Disintegrin and Metalloproteinases (ADAMs), A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTSs) and astacins in physiological and pathological processes in the central nervous system (CNS). The expression and activity of metzincins are strictly controlled at different levels (e.g., through the regulation of translation, limited activation in the extracellular space, the binding of endogenous inhibitors and interactions with other proteins). Thus, unsurprising is that the dysregulation of proteolytic activity, especially the greater expression and activation of metzincins, is associated with neurodegenerative disorders that are considered synaptopathies, especially Alzheimer's disease (AD). We review current knowledge of the functions of metzincins in the development of AD, mainly the proteolytic processing of amyloid precursor protein, the degradation of amyloid β (Aβ) peptide and several pathways for Aβ clearance across brain barriers (i.e., blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB)) that contain specific receptors that mediate the uptake of Aβ peptide. Controlling the proteolytic activity of metzincins in Aβ-induced pathological changes in AD patients' brains may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Patrycja Brzdak
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Daria Nowak
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Grzegorz Wiera
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Physiology and Molecular Neurobiology, Wroclaw UniversityWroclaw, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical UniversityWroclaw, Poland
| |
Collapse
|
48
|
Glycosylation of matrix metalloproteases and tissue inhibitors: present state, challenges and opportunities. Biochem J 2017; 473:1471-82. [PMID: 27234584 PMCID: PMC4888457 DOI: 10.1042/bj20151154] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
Current knowledge about the glycosylation of matrix metalloproteinases (MMPs) and the inhibitors of metalloproteinases (TIMPs) is reviewed. Whereas structural and functional aspects of the glycobiology of many MMPs is unknown, research on MMP-9 and MMP-14 glycosylation reveals important functional implications, such as altered inhibitor binding and cellular localization. This, together with the fact that MMPs contain conserved and many potential attachment sites for N-linked and O-linked oligosaccharides, proves the need for further studies on MMP glycobiology. Matrix metalloproteases (MMPs) are crucial components of a complex and dynamic network of proteases. With a wide range of potential substrates, their production and activity are tightly controlled by a combination of signalling events, zymogen activation, post-translational modifications and extracellular inhibition. Slight imbalances may result in the initiation or progression of specific disease states, such as cancer and pathological inflammation. As glycosylation modifies the structures and functions of glycoproteins and many MMPs contain N- or O-linked oligosaccharides, we examine, compare and evaluate the evidence for whether glycosylation affects MMP catalytic activity and other functions. It is interesting that the catalytic sites of MMPs do not contain O-linked glycans, but instead possess a conserved N-linked glycosylation site. Both N- and O-linked oligosaccharides, attached to specific protein domains, endow these domains with novel functions such as the binding to lectins, cell-surface receptors and tissue inhibitors of metalloproteases (TIMPs). Validated glycobiological data on N- and O-linked oligosaccharides of gelatinase B/MMP-9 and on O-linked structures of membrane-type 1 MMP/MMP-14 indicate that in-depth research of other MMPs may yield important insights, e.g. about subcellular localizations and functions within macromolecular complexes.
Collapse
|
49
|
Lu L, Li X, Xu P, Zheng Y, Wang X. Tenuigenin down-regulates the release of nitric oxide, matrix metalloproteinase-9 and cytokines from lipopolysaccharide-stimulated microglia. Neurosci Lett 2017; 650:82-88. [PMID: 28392358 DOI: 10.1016/j.neulet.2017.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/15/2017] [Accepted: 04/01/2017] [Indexed: 01/24/2023]
Abstract
Tenuigenin (TEN), an active component of Polygala tenuifolia root extracts, has been shown to provide neuroprotection in neurodegenerative disorders. To date, most of these studies have focused on the effect that TEN has on neurons. Because activated microglia can release neurotoxic factors that cause neuronal damage, the present study was designed to investigate the effects of TEN on activated microglia. The results showed that TEN can significantly decrease the release of nitric oxide (NO) from lipopolysaccharide (LPS)-activated rat microglia in a dose-dependent manner. The western blotting results showed that TEN did not inhibit iNOS expression at protein level. However, the electron paramagnetic resonance (EPR) technique revealed that TEN directly scavenged the NO radical. Additionally, TEN can significantly decrease the secretion and mRNA levels of matrix metalloproteinase-9 (MMP-9) and pro-inflammatory cytokines (TNF-α/IL-1β) in activated microglia. At a high dose (10-4M), TEN can significantly inhibit the secretion of another gelatinolytic MMP, MMP-2, but it had no effect on the mRNA level of MMP-2. In conclusion, these results suggest that TEN exerts an anti-inflammatory effect by down-regulating the release of NO, MMP-9 and cytokines.
Collapse
Affiliation(s)
- Li Lu
- Department of Pharmacology, Capital Medical University, Beijing 100069, PR China
| | - Xiaorong Li
- Department of Pharmacology, Capital Medical University, Beijing 100069, PR China
| | - Pingxiang Xu
- Department of Pharmacology, Capital Medical University, Beijing 100069, PR China
| | - Yan Zheng
- Department of Physiology, Capital Medical University, Beijing 100069, PR China
| | - Xiaomin Wang
- Department of Neurobiology, Capital Medical University, Beijing 100069, PR China; Beijing Institute for Brain Disorders, Beijing 100069, PR China.
| |
Collapse
|
50
|
Gentile E, Liuzzi GM. Marine pharmacology: therapeutic targeting of matrix metalloproteinases in neuroinflammation. Drug Discov Today 2016; 22:299-313. [PMID: 27697495 DOI: 10.1016/j.drudis.2016.09.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/18/2016] [Accepted: 09/26/2016] [Indexed: 01/08/2023]
Abstract
Alterations in matrix metalloproteinase (MMP) expression and activity are recognized as key pathogenetic events in several neurological disorders. This evidence makes MMPs possible therapeutic targets. The search for substances that can inhibit MMPs is moving progressively toward the screening of natural products. In particular, marine bioprospecting could be promising for the discovery of marine natural products with anti-MMP activities. Despite recent advances in this field, the possibility of using marine MMP inhibitors (MMPIs) for the treatment of neuroinflammation is still under-investigated. Here, we review the latest findings in this promising research field and the potential that marine MMPIs can have in the management and treatment of various neurological diseases.
Collapse
Affiliation(s)
- Eugenia Gentile
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy
| | - Grazia M Liuzzi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Via Orabona 4, 70125 Bari, Italy.
| |
Collapse
|