1
|
Wu A, He Y, Zhou H, Huang N, Xu H, Xia J, Zengbo L, Huang M. Downregulation of MMP-9 by epicatechin can improve the radiosensitivity of non-small cell lung cancer. J Cancer Res Ther 2024; 20:1284-1292. [PMID: 39206990 DOI: 10.4103/jcrt.jcrt_1941_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 06/03/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AND PURPOSE Radiation therapy is a crucial treatment for nonsmall cell lung cancer (NSCLC), but its effectiveness is limited by the resistance of tumor cells to radiation. This study aimed to evaluate the effect of epicatechin (EC) on radiosensitivity in NSCLC and to determine its relationships with matrix metalloproteinase (MMP)-9. METHODS MMP-9 expression was detected by Western blotting, and the expression of the DNA damage marker protein was detected by immunofluorescence. Cell viability was assessed using the CCK-8 assay, and cell proliferation was evaluated using the clonogenesis assay. Flow cytometry was used to determine the cell apoptosis, whereas cell migration and invasion were detected using the transwell assays. The cells were treated with ionizing radiation (IR) and EC to verify the sensitizing effect of EC on radiation therapy. RESULTS MMP-9 expression was elevated in the NSCLC cells and tissues. DNA damage and cell apoptosis were increased, whereas cell vigor, proliferation, migration, and invasion were significantly decreased after IR. MMP-9 knockdown strengthened the impact of IR on the biological behaviors of the cells. EC + IR had the best effect on promoting DNA damage and the biological behaviors of the NSCLC cells; alternatively, the overexpression of MMP-9 weakened the role of EC. CONCLUSIONS This study shows that EC can downregulate MMP-9 expression, promote DNA damage, reduce cell viability, proliferation, migration, and invasion, and facilitate cell apoptosis, thus, showing potential as a radiosensitizer for NSCLC.
Collapse
Affiliation(s)
- Anao Wu
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Yongmei He
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Huahua Zhou
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Nan Huang
- Department of Pulmonary, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Hongying Xu
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Jie Xia
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Lv Zengbo
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Meifang Huang
- Department of Oncology, The First People's Hospital of Qujing/The Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| |
Collapse
|
2
|
Wu X, Zhang T, Jia J, Chen Y, Zhang Y, Fang Z, Zhang C, Bai Y, Li Z, Li Y. Perspective insights into versatile hydrogels for stroke: From molecular mechanisms to functional applications. Biomed Pharmacother 2024; 173:116309. [PMID: 38479180 DOI: 10.1016/j.biopha.2024.116309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 03/27/2024] Open
Abstract
As the leading killer of life and health, stroke leads to limb paralysis, speech disorder, dysphagia, cognitive impairment, mental depression and other symptoms, which entail a significant financial burden to society and families. At present, physiology, clinical medicine, engineering, and materials science, advanced biomaterials standing on the foothold of these interdisciplinary disciplines provide new opportunities and possibilities for the cure of stroke. Among them, hydrogels have been endowed with more possibilities. It is well-known that hydrogels can be employed as potential biosensors, medication delivery vectors, and cell transporters or matrices in tissue engineering in tissue engineering, and outperform many traditional therapeutic drugs, surgery, and materials. Therefore, hydrogels become a popular scaffolding treatment option for stroke. Diverse synthetic hydrogels were designed according to different pathophysiological mechanisms from the recently reported literature will be thoroughly explored. The biological uses of several types of hydrogels will be highlighted, including pro-angiogenesis, pro-neurogenesis, anti-oxidation, anti-inflammation and anti-apoptosis. Finally, considerations and challenges of using hydrogels in the treatment of stroke are summarized.
Collapse
Affiliation(s)
- Xinghan Wu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Jia
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yining Chen
- Key laboratory for Leather Chemistry and Engineering of the Education Ministry, Sichuan University, Chengdu, Sichuan 610065, China
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenwei Fang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Bai
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhengjun Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
3
|
Rashid ZA, Bardaweel SK. Novel Matrix Metalloproteinase-9 (MMP-9) Inhibitors in Cancer Treatment. Int J Mol Sci 2023; 24:12133. [PMID: 37569509 PMCID: PMC10418771 DOI: 10.3390/ijms241512133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Matrix metalloproteinases (MMPs) belong to a family of zinc-dependent proteolytic metalloenzymes. MMP-9, a member of the gelatinase B family, is characterized as one of the most intricate MMPs. The crucial involvement of MMP-9 in extracellular matrix (ECM) remodeling underscores its significant correlation with each stage of cancer pathogenesis and progression. The design and synthesis of MMP-9 inhibitors is a potentially attractive research area. Unfortunately, to date, there is no effective MMP-9 inhibitor that passes the clinical trials and is approved by the FDA. This review primarily focuses on exploring the diverse strategies employed in the design and advancement of MMP-9 inhibitors, along with their anticancer effects and selectivity. To illuminate the essential structural characteristics necessary for the future design of novel MMP-9 inhibitors, the current narrative review highlights several recently discovered MMP-9 inhibitors exhibiting notable selectivity and potency.
Collapse
Affiliation(s)
| | - Sanaa K. Bardaweel
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
4
|
Fang J, Wang Z, Miao CY. Angiogenesis after ischemic stroke. Acta Pharmacol Sin 2023; 44:1305-1321. [PMID: 36829053 PMCID: PMC10310733 DOI: 10.1038/s41401-023-01061-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/01/2023] [Indexed: 02/26/2023] Open
Abstract
Owing to its high disability and mortality rates, stroke has been the second leading cause of death worldwide. Since the pathological mechanisms of stroke are not fully understood, there are few clinical treatment strategies available with an exception of tissue plasminogen activator (tPA), the only FDA-approved drug for the treatment of ischemic stroke. Angiogenesis is an important protective mechanism that promotes neural regeneration and functional recovery during the pathophysiological process of stroke. Thus, inducing angiogenesis in the peri-infarct area could effectively improve hemodynamics, and promote vascular remodeling and recovery of neurovascular function after ischemic stroke. In this review, we summarize the cellular and molecular mechanisms affecting angiogenesis after cerebral ischemia registered in PubMed, and provide pro-angiogenic strategies for exploring the treatment of ischemic stroke, including endothelial progenitor cells, mesenchymal stem cells, growth factors, cytokines, non-coding RNAs, etc.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
5
|
Somasundaram DB, Aravindan S, Major R, Natarajan M, Aravindan N. MMP-9 reinforces radiation-induced delayed invasion and metastasis of neuroblastoma cells through second-signaling positive feedback with NFκB via both ERK and IKK activation. Cell Biol Toxicol 2023; 39:1053-1076. [PMID: 34626302 DOI: 10.1007/s10565-021-09663-4] [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: 03/09/2021] [Accepted: 09/28/2021] [Indexed: 10/20/2022]
Abstract
Neuroblastoma (NB) progression is branded with hematogenous metastasis and frequent relapses. Despite intensive multimodal clinical therapy, outcomes for patients with progressive disease remain poor, with negligible long-term survival. Therefore, understanding the acquired molecular rearrangements in NB cells with therapy pressure and developing improved therapeutic strategies is a critical need to improve the outcomes for high-risk NB patients. We investigated the rearrangement of MMP9 in NB with therapy pressure, and unveiled the signaling that facilitates NB evolution. Radiation-treatment (RT) significantly increased MMP9 expression/activity, and the induced enzyme activity was persistently maintained across NB cell lines. Furthermore, RT-triggered NFκB transcriptional activity and this RT-induced NFκB were required/adequate for MMP9 maintenance. RT-triggered NFκB-dependent MMP9 actuated a second-signaling feedback to NFκB, facilitating a NFκB-MMP9-NFκB positive feedback cycle (PFC). Critically, MMP9-NFκB feedback is mediated by MMP9-dependent activation of IKKβ and ERK phosphotransferase activity. Beyond its tumor invasion/metastasis function, PFC-dependent MMP9 lessens RT-induced apoptosis and favors survival pathway through the activation of NFκB signaling. In addition, PFC-dependent MMP9 regulates 19 critical molecular determinants that play a pivotal role in tumor evolution. Interestingly, seven of 19 genes possess NFκB-binding sites, demonstrating that MMP9 regulates these molecules by activating NFκB. Collectively, these data suggest that RT-triggered NFκB-dependent MMP9 actuates feedback to NFκB though IKKβ- and ERK1/2-dependent IκBα phosphorylation. This RT-triggered PFC prompts MMP9-dependent survival advantage, tumor growth, and dissemination. Targeting therapy-pressure-driven PFC and/or selective inhibition of MMP9 maintenance could serve as promising therapeutic strategies for treatment of progressive NB.
Collapse
Affiliation(s)
- Dinesh Babu Somasundaram
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, BMSB 311, 940 Stanton L. Young Boulevard, Oklahoma City, OK, 73104, USA
| | | | - Ryan Major
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, BMSB 311, 940 Stanton L. Young Boulevard, Oklahoma City, OK, 73104, USA
| | - Mohan Natarajan
- Department of Pathology & Laboratory Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX, USA
| | - Natarajan Aravindan
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, BMSB 311, 940 Stanton L. Young Boulevard, Oklahoma City, OK, 73104, USA.
- Stephenson Cancer Center, Oklahoma City, OK, USA.
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Anesthesiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
6
|
Zhu MM, Ma Y, Tang M, Pan L, Liu WL. Hypoxia-induced upregulation of matrix metalloproteinase 9 increases basement membrane degradation by downregulating collagen type IV alpha 1 chain. Physiol Res 2022. [DOI: 10.33549/physiolres.934930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Hypoxia can cause basement membrane (BM) degradation in tissues. Matrix metalloproteinase 9 (MMP-9) is involved in various human cancers as well as BM degradation by downregulating type IV collagen (COL4). This study investigated the role of MMP-9 in hypoxia-mediated BM degradation in rat bone marrow based on its regulation of collagen type IV alpha 1 chain (COL4A1). Eighty male rats were randomly divided into four groups based on exposure to hypoxic conditions at a simulated altitude of 7,000 m, control (normoxia) and 3, 7, and 10 days of hypoxia exposure. BM degradation in bone marrow was determined by transmission electron microscopy. MMP-9 levels were assessed by western blot and real-time PCR, and COL4A1 levels were assessed by western blot and immunohistochemistry. Microvessels BMs in bone marrow exposed to acute hypoxia were observed by electron microscopy. MMP-9 expression increased, COL4A1 protein expression decreased, and BM degradation occurred in the 10-, 7-, and 3-day hypoxia groups compared with that in the control group (all P < 0.05). Hypoxia increased MMP-9 levels, which in turn downregulated COL4A1, thereby increasing BM degradation. MMP-9 upregulation significantly promoted BM degradation and COL4A1 downregulation. Our results suggest that MMP-9 is related to acute hypoxia-induced BM degradation in bone marrow by regulating COL4A1.
Collapse
Affiliation(s)
| | | | | | | | - WL Liu
- Affiliated Hospital of Qinghai University, Xining 810001, China;
| |
Collapse
|
7
|
Clément V, Roy V, Paré B, Goulet CR, Deschênes LT, Berthod F, Bolduc S, Gros-Louis F. Tridimensional cell culture of dermal fibroblasts promotes exosome-mediated secretion of extracellular matrix proteins. Sci Rep 2022; 12:19786. [PMID: 36396670 PMCID: PMC9672399 DOI: 10.1038/s41598-022-23433-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Extracellular matrix (ECM) secretion, deposition and assembly are part of a whole complex biological process influencing the microenvironment and other cellular behaviors. Emerging evidence is attributing a significant role to extracellular vesicles (EVs) and exosomes in a plethora of ECM-associated functions, but the role of dermal fibroblast-derived EVs in paracrine signalling is yet unclear. Herein, we investigated the effect of exosomes isolated from stimulated human dermal fibroblasts. We report that tridimensional (3D) cell culture of dermal fibroblasts promotes secretion of exosomes carrying a large quantity of proteins involved in the formation, organisation and remodelling of the ECM. In our 3D model, gene expression was highly modulated and linked to ECM, cellular migration and proliferation, as well as inflammatory response. Mass spectrometry analysis of exosomal proteins, isolated from 3D cultured fibroblast-conditioned media, revealed ECM protein enrichment, of which many were associated with the matrisome. We also show that the cytokine interleukin 6 (IL-6) is predicted to be central to the signalling pathways related to ECM formation and contributing to cell migration and proliferation. Overall, our data suggest that dermal fibroblast-derived EVs participate in many steps of the establishment of dermis's ECM.
Collapse
Affiliation(s)
- Vincent Clément
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Vincent Roy
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Bastien Paré
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Cassandra R. Goulet
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Lydia Touzel Deschênes
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - François Berthod
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - Stéphane Bolduc
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| | - François Gros-Louis
- grid.23856.3a0000 0004 1936 8390Department of Surgery, Faculty of Medicine, Laval University, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390Division of Regenerative Medicine, Laval University Experimental Organogenesis Research Center/LOEX, CHU de Québec Research Center – Enfant-Jésus Hospital, Québec, QC Canada
| |
Collapse
|
8
|
ZHU MM, MA Y, TANG M, PAN L, LIU WL. Hypoxia-induced upregulation of matrix metalloproteinase 9 increases basement membrane degradation by downregulating collagen type IV alpha 1 chain. Physiol Res 2022; 71:825-834. [PMID: 36281728 PMCID: PMC9814978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Hypoxia can cause basement membrane (BM) degradation in tissues. Matrix metalloproteinase 9 (MMP-9) is involved in various human cancers as well as BM degradation by downregulating type IV collagen (COL4). This study investigated the role of MMP-9 in hypoxia-mediated BM degradation in rat bone marrow based on its regulation of collagen type IV alpha 1 chain (COL4A1). Eighty male rats were randomly divided into four groups based on exposure to hypoxic conditions at a simulated altitude of 7,000 m, control (normoxia) and 3, 7, and 10 days of hypoxia exposure. BM degradation in bone marrow was determined by transmission electron microscopy. MMP-9 levels were assessed by western blot and real-time PCR, and COL4A1 levels were assessed by western blot and immunohistochemistry. Microvessels BMs in bone marrow exposed to acute hypoxia were observed by electron microscopy. MMP-9 expression increased, COL4A1 protein expression decreased, and BM degradation occurred in the 10-, 7-, and 3-day hypoxia groups compared with that in the control group (all P < 0.05). Hypoxia increased MMP-9 levels, which in turn downregulated COL4A1, thereby increasing BM degradation. MMP-9 upregulation significantly promoted BM degradation and COL4A1 downregulation. Our results suggest that MMP-9 is related to acute hypoxia-induced BM degradation in bone marrow by regulating COL4A1.
Collapse
Affiliation(s)
- Ming-Ming ZHU
- Affiliated Hospital of Qinghai University, Xining, China
| | - Yi MA
- Qinghai University, Xining, China,Qinghai University High Altitude Medicine Research Center, Key Laboratory of High-Altitude Medicine Ministry of Education Qinghai Provincial Key Laboratory of Plateau Medicine Application Basics Xining, China
| | - Meng TANG
- The First People’s Hospital of Yibin, Yibin, China
| | - Li PAN
- Xi’an Daxing Hospital, Xi’an, China
| | | |
Collapse
|
9
|
Mathpal S, Sharma P, Joshi T, Pande V, Mahmud S, Jeong MK, Obaidullah AJ, Chandra S, Kim B. Identification of Zinc-Binding Inhibitors of Matrix Metalloproteinase-9 to Prevent Cancer Through Deep Learning and Molecular Dynamics Simulation Approach. Front Mol Biosci 2022; 9:857430. [PMID: 35463960 PMCID: PMC9024349 DOI: 10.3389/fmolb.2022.857430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
The overexpression of matrix metalloproteinase-9 (MMP-9) is associated with tumor development and angiogenesis, and hence, it has been considered an attractive drug target for anticancer therapy. To assist in drug design endeavors for MMP-9 targets, an in silico study was presented to investigate whether our compounds inhibit MMP-9 by binding to the catalytic domain, similar to their inhibitor or not. For that, in the initial stage, a deep-learning algorithm was used for the predictive modeling of the CHEMBL321 dataset of MMP-9 inhibitors. Several regression models were built and evaluated based on R2, MAE MSE, RMSE, and Loss. The best model was utilized to screen the drug bank database containing 9,102 compounds to seek novel compounds as MMP-9 inhibitors. Then top high score compounds were selected for molecular docking based on the comparison between the score of the reference molecule. Furthermore, molecules having the highest docking scores were selected, and interaction mechanisms with respect to S1 pocket and catalytic zinc ion of these compounds were also discussed. Those compounds, involving binding to the catalytic zinc ion and the S1 pocket of MMP-9, were considered preferentially for molecular dynamics studies (100 ns) and an MM-PBSA (last 30 ns) analysis. Based on the results, we proposed several novel compounds as potential candidates for MMP-9 inhibition and investigated their binding properties with MMP-9. The findings suggested that these compounds may be useful in the design and development of MMP-9 inhibitors in the future.
Collapse
Affiliation(s)
- Shalini Mathpal
- Department of Biotechnology, Kumaun University Uttarakhand, Bhimtal, India
| | - Priyanka Sharma
- Department of Botany, DSB, Campus, Kumaun University, Nainital, India
| | - Tushar Joshi
- Department of Biotechnology, Kumaun University Uttarakhand, Bhimtal, India
| | - Veena Pande
- Department of Biotechnology, Kumaun University Uttarakhand, Bhimtal, India
| | - Shafi Mahmud
- Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
- Department of Genome Science, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Mi-Kyung Jeong
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Ahmad J. Obaidullah
- Drug Exploration and Development Chair (DEDC), Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Subhash Chandra
- Computational Biology and Biotechnology Laboratory, Department of Botany, Soban Singh Jeena University, Almora, India
- *Correspondence: Subhash Chandra, ; Bonglee Kim,
| | - Bonglee Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Subhash Chandra, ; Bonglee Kim,
| |
Collapse
|
10
|
Tian K, Du G, Wang X, Wu X, Li L, Liu W, Wu R. MMP-9 secreted by M2-type macrophages promotes Wilms' tumour metastasis through the PI3K/AKT pathway. Mol Biol Rep 2022; 49:3469-3480. [PMID: 35107742 DOI: 10.1007/s11033-022-07184-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/20/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Wilms' tumour (WT) is a malignant tumour of childhood with the typical symptoms of an abdominal mass. Tumour-associated macrophages (TAMs) accumulate and imply a poor prognosis in WT, but the mechanism of how TAMs affect the prognosis has not been fully elucidated. In this study, we aimed to present the molecular mechanisms underlying the protumorigenic capacities of TAMs in WT. METHODS TAMs were polarized into M1- and M2-type macrophages. The two types of macrophages were cocultured with SK-NEP-1 cells, and their cell viability and invasion ability were measured. Matrix metalloproteinase 9 (MMP9) expression was assessed in different types of macrophages, and the role of MMP9 in WT was explored. Then data from children diagnosed with WT in our department between February 2006 and July 2014 were retrospectively analysed, the tumour tissues were analysed to explore the distribution of MMP9. Kaplan-Meier analysis of the relationship between MMP9 expression and follow-up information was performed. RESULTS The results showed that M2-type macrophages could improve the viability and invasive ability of SK-NEP-1 cells. MMP9 expression in M2-type macrophages was significantly higher than that in M1-type macrophages. MMP9 could activate the AKT/PI3K signalling pathway to initiate the epithelial-mesenchymal transition (EMT) process, and promote the proliferation and invasion of WT. In WT tissue, the MMP9 expression level was elevated and it was located in the tumour stroma, which was the same as M2-type macrophage location, and a high level of MMP9 predicted poor survival. CONCLUSION M2-type macrophages facilitate tumour proliferation and metastasis by secreting MMP9 to enhance the EMT process via a PI3K/AKT dependent pathway in Wilms' tumour.
Collapse
Affiliation(s)
- Kaixuan Tian
- Department of Pediatric Orthopaedics, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, People's Republic of China.,Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Guoqiang Du
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Xiaoqing Wang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Xiangyu Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Long Li
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China
| | - Wei Liu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China.
| | - Rongde Wu
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, People's Republic of China.
| |
Collapse
|
11
|
Liu N, Wang X, Wu H, Lv X, Xie H, Guo Z, Wang J, Dou G, Zhang C, Sun M. Computational study of effective matrix metalloproteinase 9 (MMP9) targeting natural inhibitors. Aging (Albany NY) 2021; 13:22867-22882. [PMID: 34607974 PMCID: PMC8544340 DOI: 10.18632/aging.203581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/10/2021] [Indexed: 02/06/2023]
Abstract
Object: The present study screened ideal lead natural compounds that could target and inhibit matrix metalloproteinase 9 (MMP9) protein from the ZINC database to develop drugs for clear cell renal cell carcinoma (CCRCC)-targeted treatment. Methods: Discovery Studio 4.5 was used to compare and screen the ligands with the reference drug, solasodine, to identify ideal candidate compounds that could inhibit MMP9. The LibDock module was used to analyze compounds that could strongly bind to MMP9, and the top 20 compounds determined by the LibDock score were selected for further research. ADME and TOPKAT modules were used to choose the safe compounds from these 20 compounds. The selected compounds were analyzed using the CDOCKER module for molecular docking and feature mapping for pharmacophore prediction. The stability of these compound–MMP9 complexes was analyzed by molecular dynamic simulation. Cell counting kit-8, colony-forming, and scratch assays were used to analyze the anti-CCRCC effects of these ligands. Results: Strong binding to MMP9 was exhibited by 6,762 ligands. Among the top 20 compounds, sappanol and sventenin exhibited nearly undefined blood–brain barrier level and lower aqueous solubility, carcinogenicity, and hepatotoxicity than the positive control drug, solasodine. Additionally, these compounds exhibited lower potential energies with MMP9, and the ligand–MMP9 complexes were stable in the natural environment. Furthermore, sappanol inhibited CCRCC cell migration and proliferation. Conclusion: Sappanol and sventenin are safe and reliable compounds to target and inhibit MMP9. Sappanol can CCRCC cell migration and proliferation. These two compounds may give new thought to the targeted therapy for patients with CCRCC.
Collapse
Affiliation(s)
- Naimeng Liu
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xinhui Wang
- Department of Oncology, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Xiaye Lv
- Department of Hematology, The First Clinical Medical School of Lanzhou University, Lanzhou, Gansu, China
| | - Haoqun Xie
- Clinical College, Jilin University, Changchun, China
| | - Zhen Guo
- Clinical College, Jilin University, Changchun, China
| | - Jing Wang
- Clinical College, Jilin University, Changchun, China
| | - Gaojing Dou
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China.,Clinical College, Jilin University, Changchun, China
| | - Chenxi Zhang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Mindan Sun
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Chaves Filho AJM, Mottin M, Soares MVR, Jucá PM, Andrade CH, Macedo DS. Tetracyclines, a promise for neuropsychiatric disorders: from adjunctive therapy to the discovery of new targets for rational drug design in psychiatry. Behav Pharmacol 2021; 32:123-141. [PMID: 33595954 DOI: 10.1097/fbp.0000000000000585] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Major mental disorders, such as schizophrenia, bipolar disorder, and major depressive disorder, represent the leading cause of disability worldwide. Nevertheless, the current pharmacotherapy has several limitations, and a large portion of patients do not respond appropriately to it or remain with disabling symptoms overtime. Traditionally, pharmacological interventions for psychiatric disorders modulate dysfunctional neurotransmitter systems. In the last decades, compelling evidence has advocated for chronic inflammatory mechanisms underlying these disorders. Therefore, the repurposing of anti-inflammatory agents has emerged as an attractive therapeutic tool for mental disorders. Minocycline (MINO) and doxycycline (DOXY) are semisynthetic second-generation tetracyclines with neuroprotective and anti-inflammatory properties. More recently, the most promising results obtained in clinical trials using tetracyclines for major psychiatric disorders were for schizophrenia. In a reverse translational approach, tetracyclines inhibit microglial reactivity and toxic inflammation by mechanisms related to the inhibition of nuclear factor kappa B signaling, cyclooxygenase 2, and matrix metalloproteinases. However, the molecular mechanism underlying the effects of these tetracyclines is not fully understood. Therefore, the present review sought to summarize the latest findings of MINO and DOXY use for major psychiatric disorders and present the possible targets to their molecular and behavioral effects. In conclusion, tetracyclines hold great promise as (ready-to-use) agents for being used as adjunctive therapy for human neuropsychiatric disorders. Hence, the understanding of their molecular mechanisms may contribute to the discovery of new targets for the rational drug design of novel psychoactive agents.
Collapse
Affiliation(s)
- Adriano José Maia Chaves Filho
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
- Laboratory for Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO
| | - Melina Mottin
- Laboratory for Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO
| | - Michele Verde-Ramo Soares
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
| | - Paloma Marinho Jucá
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Drug Design, LabMol, Faculdade de Farmácia, Faculty of Pharmacy, Universidade Federal de Goiás, Goiânia, GO
| | - Danielle S Macedo
- Neuropharmacology Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil
| |
Collapse
|
13
|
Valproate Sodium Protects Blood Brain Barrier Integrity in Intracerebral Hemorrhage Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8884320. [PMID: 33224434 PMCID: PMC7676278 DOI: 10.1155/2020/8884320] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/28/2020] [Accepted: 10/15/2020] [Indexed: 01/04/2023]
Abstract
Valproate sodium (VPA) is a traditional antiepileptic drug with a neuroprotective role in cerebrovascular disease. After intracerebral hemorrhage (ICH), mechanical compression by hematoma, neuroinflammation, oxidative stress, and cytotoxicity of hematoma lysates caused the destruction of the blood brain barrier (BBB). Targeting BBB is a major therapeutic method for patients with ICH. The purpose of the present study was to explore the role of VPA in preserving BBB integrity in the ICH model and investigate the underlying molecular mechanisms. One hundred and thirty-six adult male CD1 mice were randomly divided into five groups in the study. Mice subjected to ICH were administered intraperitoneally with VPA at 3, 24, and 48 h post-ICH, respectively. Neurobehavioral assessments, BBB permeability, Evans blue fluorescence, hematoma volume, and protein expression were evaluated. The administration of VPA reduced BBB permeability and improved the neurobehavior significantly post-ICH. VPA administration significantly decreased the expression of phosphorylated nuclear factor-kappa B (p-NFκB), matrix metalloproteinases 9 (MMP9), tumor necrosis factorα (TNFα), and interleukin-6 (IL-6), while it enhanced the expression of claudin 5 and occludin in the brain. In conclusion, VPA administration maintained the integrity of BBB after experimental ICH, thus reducing brain edema and improving the neurological outcomes. Therefore, VPA administration might be a new therapeutic method to protect BBB integrity for patients with ICH.
Collapse
|
14
|
Yang C, Yang Y, DeMars KM, Rosenberg GA, Candelario-Jalil E. Genetic Deletion or Pharmacological Inhibition of Cyclooxygenase-2 Reduces Blood-Brain Barrier Damage in Experimental Ischemic Stroke. Front Neurol 2020; 11:887. [PMID: 32973660 PMCID: PMC7468510 DOI: 10.3389/fneur.2020.00887] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022] Open
Abstract
Cyclooxygenase (COX)-2 and matrix metalloproteinase (MMP)-9 are two crucial mediators contributing to blood-brain barrier (BBB) damage during cerebral ischemia. However, it is not known whether MMP-9 activation is involved in COX-2-mediated BBB disruption in ischemic stroke. In this study, we hypothesized that genetic deletion or pharmacological inhibition of COX-2 reduces BBB damage by reducing MMP-9 activity in a mouse model of ischemic stroke. Male COX-2 knockout (COX-2-/-) and wild-type (WT) mice were subjected to 60 min of middle cerebral artery occlusion (MCAO) followed by 24 h of reperfusion. Genetic deletion of COX-2 or post-ischemic treatment with CAY10404, a highly selective COX-2 inhibitor, significantly reduced BBB damage and hemorrhagic transformation, as assessed by immunoglobulin G (IgG) extravasation and brain hemoglobin (Hb) levels, respectively. Immunoblotting analysis showed that tight junction proteins (TJPs) zonula occludens (ZO)-1 and occludin as well as junctional adhesion molecule-A (JAM-A) and the basal lamina protein collagen IV were dramatically reduced in the ischemic brain. Stroke-induced loss of these BBB structural proteins was significantly attenuated in COX-2-/- mice. Similarly, stroke-induced loss of ZO-1 and occludin was significantly attenuated by CAY10404 treatment. Ischemia-induced increase in MMP-9 protein levels in the ipsilateral cerebral cortex was significantly reduced in COX-2-/- mice. Stroke induced a dramatic increase in MMP-9 enzymatic activity in the ischemic cortex, which was markedly reduced by COX-2 gene deficiency or pharmacological inhibition with CAY10404. Levels of myeloperoxidase (MPO, an indicator of neutrophil infiltration into the brain parenchyma), neutrophil elastase (NE), and lipocalin-2 (LCN2, also known as neutrophil gelatinase-associated lipocalin), measured by western blot and specific ELISA kits, respectively, were markedly increased in the ischemic brain. Increased levels of markers for neutrophil infiltration were significantly reduced in COX-2-/- mice compared with WT controls following stroke. Altogether, neurovascular protective effects of COX-2 blockade are associated with reduced BBB damage, MMP-9 expression/activity and neutrophil infiltration. Our study shows for the first time that MMP-9 is an important downstream effector contributing to COX-2-mediated neurovascular damage in ischemic stroke. Targeting the COX-2/MMP-9 pathway could represent a promising strategy to reduce neuroinflammatory events in order to preserve the BBB integrity and ameliorate ischemic stroke injury.
Collapse
Affiliation(s)
- Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Yi Yang
- Department of Neurology, Center for Memory and Aging, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Kelly M DeMars
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Gary A Rosenberg
- Department of Neurology, Center for Memory and Aging, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Chronic Hypoxia-Induced Microvessel Proliferation and Basal Membrane Degradation in the Bone Marrow of Rats Regulated through the IL-6/JAK2/STAT3/MMP-9 Pathway. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9204708. [PMID: 32047820 PMCID: PMC7003287 DOI: 10.1155/2020/9204708] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/08/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022]
Abstract
Chronic hypoxia (CH) is characterized by long-term hypoxia that is associated with microvessel proliferation and basal membrane (BM) degradation in tissues. The IL-6/JAK2/STAT3/MMP-9 pathway has been described in a variety of human cancers and plays an essential role in microvessel proliferation and BM degradation. Therefore, this study investigated the role of the IL-6/JAK2/STAT3/MMP-9 pathway in hypoxia-mediated microvessel proliferation and BM degradation in the rat bone marrow. Eighty pathogen-free Sprague Dawley male rats were randomly divided into four groups (20 per group)—control group, CH group (exposed to hypoxia in a hypobaric chamber at a simulated altitude of 5000 m for 28 d), CH + STAT3 inhibitor group (7.5 mg/kg/d), and CH + DMSO group. Microvessel density (MVD) and BM degradation in the bone marrow were determined by immunofluorescence staining and transmission electron microscopy. Serum IL-6 levels were assessed by enzyme-linked immunosorbent assay (ELISA), and the levels of P-JAK2, P-STAT3, and MMP-9 were assessed by western blot analysis and real-time reverse transcription PCR (RT-PCR). Hypoxia increased serum IL-6 levels, which in turn increased JAK2 and STAT3 phosphorylation, which subsequently upregulated MMP-9. Overexpression of MMP-9 significantly promoted the elevation of MVD and BM degradation. Inhibition of STAT3 using an inhibitor, SH-4-54, significantly downregulated MMP-9 expression and decreased MVD and BM degradation. Surprisingly, STAT3 inhibition also decreased serum IL-6 levels and JAK2 phosphorylation. Our results suggest that the IL-6/JAK2/STAT3/MMP-9 pathway might be related to CH-induced microvessel proliferation and BM degradation in the bone marrow.
Collapse
|
16
|
Yi X, Sui G, Zhou Q, Wang C, Lin J, Chai Z, Zhou J. Variants in matrix metalloproteinase-9 gene are associated with hemorrhagic transformation in acute ischemic stroke patients with atherothrombosis, small artery disease, and cardioembolic stroke. Brain Behav 2019; 9:e01294. [PMID: 31074588 PMCID: PMC6576165 DOI: 10.1002/brb3.1294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/29/2019] [Accepted: 04/06/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The potential effect of matrix metalloproteinase-9 (MMP-9) variants and these variants interactions on hemorrhagic transformation (HT) risk after ischemic stroke (IS) remain unclear. The aims of present study were to investigate the associations of six variants in MMP-9 with HT, and these variants interactions whether related to increased HT risk. METHOD A total of 705 patients with IS who were admitted to the participating hospitals within 48 hr of symptom onset were consecutively enrolled between March 2014 and December 2016. HT was confirmed by brain computed tomography (CT) scan during 14 days from stroke onset. Six variants of MMP-9 gene were measured by mass spectrometry. Interactions of gene variant-gene variant were assessed through generalized multifactor dimensionality reduction method (GMDR). RESULTS HT occurred in 104 (14.8%) patients. There were no differences in genotypes for the six variants between patients with and without HT using univariate analysis (all p > 0.05). GMDR analysis revealed that there was a synergistic effect of gene variant-gene variant interactions between rs3918242 and rs3787268 in MMP-9 gene. Cox regression analysis showed that high-risk interactions of rs3918242 and rs3787268 were associated with increased risk of HT after adjusting for covariates (hazard ratio: 2.08; 95% confidence interval: 1.34-7.85; p = 0.016). CONCLUSION Incidence of HT is common in acute IS in Chinese population. The mechanisms leading to HT are most likely multifactorial. Two-loci interactions of rs3918242 and rs3787268 in MMP-9 gene may confer a higher risk for HT.
Collapse
Affiliation(s)
- Xingyang Yi
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Guo Sui
- Nursing Department, People's Hospital of Deyang City, Deyang, China
| | - Qiang Zhou
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chun Wang
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| | - Jing Lin
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhenxiao Chai
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ju Zhou
- Department of Neurology, People's Hospital of Deyang City, Deyang, China
| |
Collapse
|
17
|
Wang Q, Wei J, Shi Y. Platelet microvesicles promote the recovery of neurological function in mouse model of cerebral infarction by inducing angiogenesis. Biochem Biophys Res Commun 2019; 513:997-1004. [PMID: 31005253 DOI: 10.1016/j.bbrc.2019.04.083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
The aim of this study is to investigate the effect of PMVs on mice with ischemic cerebral infarction and its mechanism. Male C57BL/6 mice were selected, and the right focal cortical infarction model was established via cauterization under a microscope and randomly divided into sham operation (Sham) group, normal saline control (Saline) group and platelet microvesicles intervention (PMVs) group. At 1 h after modeling, 5 μL of PMVs (50 μg/mL) or normal saline was injected into the lateral ventricle. The neurological function of mice in each group was evaluated at 1, 3, 7, 14 and 28 d after modeling. After 28 d, the cerebral infarction area was detected via 2,3,5-triphenyltetrazolium chloride (TTC) staining. At 7 and 28 d after modeling, the blood vessel density, proliferation rate of new vessels and encapsulation rate of pericytes were detected via immunofluorescence staining. Moreover, the changes in cerebral cortical blood flow at the infarction side were detected before modeling and at 7 and 28 d after modeling, respectively. Finally, the expressions of proangiogenic factors vascular endothelial growth factor (VEGF), angiopoietin-1 (Ang-1) and N-Cadherin were detected via Western blotting at 3, 7 and 28 d after modeling. PMVs could promote the improvement of neurological function and significantly reduce the cerebral infarction volume in mice with cerebral infarction. PMVs promoted proliferation of new vessels and increased blood vessel density at the infarction edge in mice with cerebral infarction. PMVs could increase the encapsulation rate of pericytes at the infarction edge and improve the permeability of blood-brain barrier in mice with cerebral infarction. PMVs could increase the cerebral cortical blood flow perfusion in mice with cerebral infarction. PMVs could increase proangiogenic factors in brain tissues in mice with cerebral infarction. PMVs could significantly improve the recovery of neurological function in mice with cerebral infarction, which is closely related to the ability of PMVs to promote angiogenesis at the infarction edge. The possible mechanism is that PMVs facilitate angiogenesis after cerebral infarction through promoting the expressions of VEGF, Ang-1 and N-Cadherin. More importantly, the new vessels promoted by PMVs have complete structure and perfect function, and can improve the cerebral blood flow perfusion at the infarction side.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, The First Hospital of Xi'an, Xi'an, China
| | - Jiachen Wei
- Department of Endocrinology, The First Hospital of Xi'an, Xi'an, China
| | - Yaling Shi
- Department of Neurology, The First Hospital of Xi'an, Xi'an, China.
| |
Collapse
|
18
|
Qing-Feng S, Ying-Peng X, Tian-Tong X. Matrix metalloproteinase-9 and p53 involved in chronic fluorosis induced blood-brain barrier damage and neurocyte changes. Arch Med Sci 2019; 15:457-466. [PMID: 30899299 PMCID: PMC6425220 DOI: 10.5114/aoms.2019.83294] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/18/2017] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION A large number of basic and clinical studies have confirmed that fluoride produces toxic effects on multiple organ systems in the body including the nervous system. MATERIAL AND METHODS One hundred twenty Wistar rats were randomly divided into 4 groups with 30 in each group: a high fluoride group (drinking 200 mg/l fluoridated water, 24 weeks); a high fluoride control group (drinking distilled water, 24 weeks); a fluoride removal group (drinking fluoridated water, 12 W; then distilled water, 12 W) and a defluorination control group (drinking distilled water, 24 weeks). RESULTS The high fluoride and fluoride removal groups had spinal cord astrocyte edema. The apoptosis rate of spinal nerve cells in the high fluoride group and fluoride removal group were significantly higher (p < 0.01) than in the fluoride control and defluorination control group. The Evans blue (EB) content, matrix metalloproteinase-9 (MMP-9) and p53 expression in the high fluoride group and fluoride removal group were higher (p < 0.01) than in the fluoride control and defluorination control group. CONCLUSIONS The apoptosis of spinal cord nerve cells is obviously higher in rats with chronic fluoride exposure. Chronic fluoride exposure leads to high expression of MMP-9, and results in increased damage of the blood-spinal cord barrier. Increased p53 may be one of the factors causing damage. Short-term removal of fluoride has no obvious recovery in apoptosis of spinal cord nerve cells; highly expressed MMP-9 and p53 may be one of the reasons for unrecovered function.
Collapse
Affiliation(s)
- Shen Qing-Feng
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Xia Ying-Peng
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Xu Tian-Tong
- Department of Spine Surgery, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
19
|
Matrix Metalloproteinase-9 (MMP-9) as a Cancer Biomarker and MMP-9 Biosensors: Recent Advances. SENSORS 2018; 18:s18103249. [PMID: 30262739 PMCID: PMC6211011 DOI: 10.3390/s18103249] [Citation(s) in RCA: 437] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/17/2022]
Abstract
As one of the most widely investigated matrix metalloproteinases (MMPs), MMP-9 is a significant protease which plays vital roles in many biological processes. MMP-9 can cleave many extracellular matrix (ECM) proteins to regulate ECM remodeling. It can also cleave many plasma surface proteins to release them from the cell surface. MMP-9 has been widely found to relate to the pathology of cancers, including but not limited to invasion, metastasis and angiogenesis. Some recent research evaluated the value of MMP-9 as biomarkers to various specific cancers. Besides, recent research of MMP-9 biosensors discovered various novel MMP-9 biosensors to detect this enzyme. In this review, some recent advances in exploring MMP-9 as a biomarker in different cancers are summarized, and recent discoveries of novel MMP-9 biosensors are also presented.
Collapse
|
20
|
Li XF, Zhang XJ, Zhang C, Wang LN, Li YR, Zhang Y, He TT, Zhu XY, Cui LL, Gao BL. Ulinastatin protects brain against cerebral ischemia/reperfusion injury through inhibiting MMP-9 and alleviating loss of ZO-1 and occludin proteins in mice. Exp Neurol 2017; 302:68-74. [PMID: 29291404 DOI: 10.1016/j.expneurol.2017.12.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND The effects of Ulinastatin (UTI) on the blood-brain barrier (BBB) in the acute phase of cerebral ischemia/reperfusion (I/R) are not clear. This study was to investigate the potential protective effects of UTI on the BBB and the underlying mechanisms. METHODS Male CD-1 mice were subjected to transient middle cerebral artery occlusion (tMCAO) and randomly assigned to four groups: Sham (sham-operated), tMCAO (tMCAO+0.9% saline), UTI-L (tMCAO+UTI 1500U/100g) and UTI-H (tMCAO+UTI 3000U/100g) group. UTI was administered immediately after reperfusion in the UTI-L and UTI-H groups. At 24h after reperfusion, the neurological deficit, brain water content, and infarct volume were determined. Western blot and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to examine the expression of matrix metalloproteinase (MMP)-9, Zonula occludens-1 (ZO-1) and occludin in ischemic cerebral cortex. The integrity of the BBB was assessed by the leakage of Evans blue. RESULTS Compared with tMCAO group, both UTI-L and UTI-H groups showed significantly (P<0.001) ameliorated the neurological deficit (2.00±0.71 and 1.60±0.55 vs. 4.60±0.55), lessened brain water content (82.99%±0.21% and 82.05%±0.59% vs. 84.28%±0.0.57%) and decreased the infarct volume (38.52%±1.72% and 24.78%±1.20% vs. 49.48%±1.93%). In addition, significantly (P<0.001) decreased expression of MMP-9 (0.48±0.06 and 0.37±0.05 vs.0.76±0.10 for protein and 2.88±0.23 and 2.17±0.16 vs. 3.90±0.24 for mRNA) and alleviated loss of ZO-1 (0.19±0.04 and 0.24±0.05 vs. 0.25±0.03) and occludin (0.74±0.08 and 0.87±0.07 vs. 0.94±0.06) proteins were observed in both UTI-L and UTI-H groups. CONCLUSION UTI protects the brain against ischemic injury potentially via down-regulating the expression of MMP-9 and alleviating loss of ZO-1 and occludin proteins to restore the BBB permeability.
Collapse
Affiliation(s)
- Xiao-Fang Li
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiang-Jian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China.
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Li-Na Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Yao-Ru Li
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Ye Zhang
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Ting-Ting He
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Xing-Yuan Zhu
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Li-Li Cui
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Diseases, Shijiazhuang, Hebei, China
| | - Bu-Lang Gao
- Department of Interventional Treatment, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
21
|
Vascular basement membrane alterations and β-amyloid accumulations in an animal model of cerebral small vessel disease. Clin Sci (Lond) 2017; 131:1001-1013. [PMID: 28348005 DOI: 10.1042/cs20170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 12/23/2022]
Abstract
Non-amyloid cerebral small vessel disease (CSVD) and cerebral amyloid angiopathy (CAA) may be interrelated through the damaged basement membranes (BMs) and extracellular matrix changes of small vessels, resulting in a failure of β-amyloid (Aβ) transport and degradation. We analyzed BM changes and the pattern of deposition of Aβ in the walls of blood vessels in spontaneously hypertensive stroke-prone rats (SHRSP), a non-transgenic CSVD model. In 45 SHRSP and 38 Wistar rats aged 18 to 32 weeks: (i) the percentage area immunostained for vascular collagen IV and laminin was quantified; (ii) the capillary BM thickness as well as endothelial and pericyte pathological changes were analysed using transmission electron microscopy (TEM); and (iii) the presence of vascular Aβ was assessed. Compared with controls, SHRSP exhibited a significantly higher percentage area immunostained with collagen IV in the striatum and thalamus. SHRSP also revealed an age-dependent increase of the capillary BM thickness and of endothelial vacuoles (caveolae) within subcortical regions. Endogenous Aβ deposits in the walls of small blood vessels were observed in the cortex (with the highest incidence found within fronto-parietal areas), striatum, thalamus and hippocampus. Vascular β-amyloid accumulations were frequently detected at sites of small vessel wall damage. Our data demonstrate changes in the expression of collagen IV and of the ultrastructure of BMs in the small vessels of SHRSP. Alterations are accompanied by vascular deposits of endogenous Aβ. Impaired β-amyloid clearance along perivascular and endothelial pathways and failure of extracellular Aβ degradation may be the key mechanisms connecting non-amyloid CSVD and CAA.
Collapse
|
22
|
Wang Y, Wang N, Cai B, Wang GY, Li J, Piao XX. In vitro model of the blood-brain barrier established by co-culture of primary cerebral microvascular endothelial and astrocyte cells. Neural Regen Res 2016; 10:2011-7. [PMID: 26889191 PMCID: PMC4730827 DOI: 10.4103/1673-5374.172320] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Drugs for the treatment and prevention of nervous system diseases must permeate the blood-brain barrier to take effect. In vitro models of the blood-brain barrier are therefore important in the investigation of drug permeation mechanisms. However, to date, no unified method has been described for establishing a blood-brain barrier model. Here, we modified an in vitro model of the blood-brain barrier by seeding brain microvascular endothelial cells and astrocytes from newborn rats on a polyester Transwell cell culture membrane with 0.4-µm pores, and conducted transepithelial electrical resistance measurements, leakage tests and assays for specific blood-brain barrier enzymes. We show that the permeability of our model is as low as that of the blood-brain barrier in vivo. Our model will be a valuable tool in the study of the mechanisms of action of neuroprotective drugs.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Ning Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Biao Cai
- Key Laboratory of Xin'an Medicine, Ministry of Education; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Guang-Yun Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Jing Li
- Key Laboratory of Xin'an Medicine, Ministry of Education; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| | - Xing-Xing Piao
- Key Laboratory of Xin'an Medicine, Ministry of Education; Institute for Pharmacodynamics and Safety Evaluation of Chinese Medicine, Anhui Academy of Chinese; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui Province, China
| |
Collapse
|
23
|
Zhang X, Cao X, Xu X, Li A, Xu Y. Correlation between the -1562C/T polymorphism in the matrix metalloproteinase-9 gene and hemorrhagic transformation of ischemic stroke. Exp Ther Med 2015; 9:1043-1047. [PMID: 25667675 PMCID: PMC4316928 DOI: 10.3892/etm.2015.2186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 12/17/2014] [Indexed: 12/04/2022] Open
Abstract
The aim of the present study was to investigate the correlation between the −1562C/T polymorphism in an intron of the matrix metalloproteinase-9 (MMP-9) gene and hemorrhagic transformation of ischemic stroke (IS). Using polymerase chain reaction-restriction fragment length polymorphism, the −1562C/T polymorphisms in 222 patients with IS were detected. The patients were divided into hemorrhagic transformation (HT; 84 cases) and non-hemorrhagic transformation (NHT) groups (138 cases) depending on the results from the susceptibility-weighted magnetic resonance imaging, which was performed between one and two weeks following stroke onset. The allele frequencies were subsequently compared. Baseline data of the two groups were comparable. The HT group exhibited a significantly lower frequency of the CT+TT genotype compared with the NHT group (17.86 vs. 30.43%, P<0.05). In addition, the frequency of T allele was significantly lower in the HT group compared with the NHT group (8.93 vs. 15.94%, P<0.05). Therefore, the results indicated that the −1562C/T polymorphism in the MMP-9 gene is correlated with hemorrhagic transformation of IS in the population studied. Furthermore, the T allele may be a protective factor for hemorrhagic transformation of IS in this population.
Collapse
Affiliation(s)
- Xiaoman Zhang
- Department of Neurology, The First People's Hospital of Zhengzhou, Zhengzhou, Henan 450004, P.R. China
| | - Xinhui Cao
- Department of Neurology, The First People's Hospital of Zhengzhou, Zhengzhou, Henan 450004, P.R. China
| | - Xiaoyu Xu
- Department of Neurology, The First People's Hospital of Zhengzhou, Zhengzhou, Henan 450004, P.R. China
| | - Aifan Li
- Department of Neurology, The First People's Hospital of Zhengzhou, Zhengzhou, Henan 450004, P.R. China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|