1
|
Ruan L, Zheng M, Xia X, Pang C, Wang Y, Fan Z, Yang J, Qing Q, Lin H, Tao Y, Wang J, Wang L. Exploring the Constituents and Mechanisms of Polygonum multiflorum Thunb. in Mitigating Ischemic Stroke: A Network Pharmacology and Molecular Docking Study. Comb Chem High Throughput Screen 2025; 28:781-797. [PMID: 38623977 DOI: 10.2174/0113862073285988240229081558] [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: 11/04/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 04/17/2024]
Abstract
BACKGOUND Polygonum multiflorum Thunb. (PMT) has shown promise in exerting cerebrovascular protective effects, and its potential for treating ischemic stroke (IS) has garnered attention. However, the lack of clarity regarding its chemical constituents and mechanisms has significantly hindered its clinical application. METHODS In this study, we employed network pharmacology and molecular docking techniques for the first time to elucidate the potential compounds and targets of PMT in treating IS. The databases CTD, DrugBank, DisGeNET, GeneCards, OMIM, TTD, PGKB, NCBI, TCMIP, CNKI, PubMed, ZINC, STITCH, BATMAN, ETCM and Swiss provided information on targets related to IS and components of PMT, along with their associated targets. We constructed "compoundtarget" and protein-protein interaction (PPI) networks sourced from the STRING database using the Cytoscape software. Gene Ontology (GO) enrichment analysis and KEGG pathway analysis were conducted using the DAVID database. Molecular docking between core targets and active compounds was conducted using Autodock4 software. Experiments were performed in an oxygen- glucose deprivation and reperfusion (OGD/R) model to validate the anti-IS activity of compounds isolated from PMT preliminarily. Network pharmacological analysis revealed 16 core compounds, including resveratrol, polydatin, TSG, ω-hydroxyemodin, emodin anthrone, tricin, moupinamide, and others, along with 11 high-degree targets, such as PTGS1, PTGS2, ADORA1, ADORA2, CA1, EGFR, ESR1, ESR2, SRC, MMP3 and MMP9. RESULTS GO and KEGG enrichment analyses revealed the involvement of HIF-1, Akt signaling pathway and energy metabolism-related signaling pathways. Molecular docking results emphasized eight key compounds and confirmed their interactions with corresponding targets. In vitro OGD/R model experiments identified TSG and tricin as the primary active substances within PMT for its anti-stroke activity. CONCLUSION This study contributes new insights into the potential development of PMT for stroke prevention and treatment.
Collapse
Affiliation(s)
- Lingyu Ruan
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Mengyun Zheng
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Xinru Xia
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Chaofan Pang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Yating Wang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Zhiwei Fan
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Jingtian Yang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Qing Qing
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Hongyan Lin
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Yuheng Tao
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| | - Junsong Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Liqun Wang
- School of Pharmacy and School of Biological and Food Engineering, Changzhou University, 21 GeHu Middle Road, Changzhou, 213164, China
| |
Collapse
|
2
|
Escolano-Lozano F, Gries E, Schlereth T, Dimova V, Baka P, Vlckova E, König S, Birklein F. Local and Systemic Expression Pattern of MMP-2 and MMP-9 in Complex Regional Pain Syndrome. THE JOURNAL OF PAIN 2021; 22:1294-1302. [PMID: 33892152 DOI: 10.1016/j.jpain.2021.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 01/28/2023]
Abstract
Matrix metalloproteinases (MMP)-2 and MMP-9 play important roles in inflammation as well as in pain processes. For this reason, we compared the concentrations of these enzymes in skin and serum of patients with complex regional pain syndrome (CRPS), other pain diseases and healthy subjects. We analyzed ipsi- and contralateral skin biopsies of 18 CRPS patients, as well as in 10 pain controls and 9 healthy subjects. Serum samples were analyzed from 20 CRPS, 17 pain controls and 17 healthy subjects. All samples were analyzed with ELISA. Concentrations were then compared to clinical data as well as to quantitative sensory testing data.MMP-2 was increased in both ipsi- and contralateral skin biopsies of CRPS patients compared to healthy subjects. While low ipsilateral MMP-2 was associated with trophic changes, contralateral MMP-2 inversely correlated with the CRPS severity. MMP-9 was also locally increased in ipsilateral CRPS skin, and higher ipsi- and contralateral MMP-9 levels correlated with CRPS severity. We conclude that MMP-2 and MMP-9 are differently expressed depending on the clinical phenotype in CRPS. PERSPECTIVE: This article describes an upregulation of MMPs in CRPS and pain controls and shows different expression of MMP-2 and -9 depending on clinical phenotype in CRPS. These results provide evidence that MMP-2 and -9 play a key role in CRPS pathophysiology.
Collapse
Affiliation(s)
| | - Eva Gries
- University Hospital Mainz, Cardiology Department, Mainz, Germany
| | - Tanja Schlereth
- DKD Helios Klinik Wiesbaden, Department of Neurology, Wiesbaden, Germany
| | - Violeta Dimova
- University Hospital Mainz, Neurology Department, Mainz, Germany
| | - Panoraia Baka
- University Hospital Mainz, Neurology Department, Mainz, Germany
| | - Eva Vlckova
- University Hospital Brno, Neurology Department, Brno, Czech Republic
| | - Simone König
- Core Unit Proteomics, Interdisciplinary Center for Clinical Research, Medical Faculty, University of Münster, Germany
| | - Frank Birklein
- University Hospital Mainz, Neurology Department, Mainz, Germany
| |
Collapse
|
3
|
Abstract
Climate change is one of the biggest challenges humanity is facing in the 21st century. Two recognized sequelae of climate change are global warming and air pollution. The gradual increase in ambient temperature, coupled with elevated pollution levels have a devastating effect on our health, potentially contributing to the increased rate and severity of numerous neurological disorders. The main aim of this review paper is to shed some light on the association between the phenomena of global warming and air pollution, and two of the most common and debilitating neurological conditions: stroke and neurodegenerative disorders. Extreme ambient temperatures induce neurological impairment and increase stroke incidence and mortality. Global warming does not participate in the etiology of neurodegenerative disorders, but it exacerbates symptoms of dementia, Alzheimer's disease (AD) and Parkinson's Disease (PD). A very close link exists between accumulated levels of air pollutants (principally particulate matter), and the incidence of ischemic rather than hemorrhagic strokes. People exposed to air pollutants have a higher risk of developing dementia and AD, but not PD. Oxidative stress, changes in cardiovascular and cerebrovascular haemodynamics, excitotoxicity, microglial activation, and cellular apoptosis, all play a central role in the overlap of the effect of climate change on neurological disorders. The complex interactions between global warming and air pollution, and their intricate effect on the nervous system, imply that future policies aimed to mitigate climate change must address these two challenges in unison.
Collapse
Affiliation(s)
- Christian Zammit
- Anatomy Department, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| | - Natalia Torzhenskaya
- Anatomy Department, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| | | | - Jean Calleja Agius
- Anatomy Department, Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| |
Collapse
|
4
|
Vancsik T, Máthé D, Horváth I, Várallyaly AA, Benedek A, Bergmann R, Krenács T, Benyó Z, Balogh A. Modulated Electro-Hyperthermia Facilitates NK-Cell Infiltration and Growth Arrest of Human A2058 Melanoma in a Xenograft Model. Front Oncol 2021; 11:590764. [PMID: 33732640 PMCID: PMC7959784 DOI: 10.3389/fonc.2021.590764] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Modulated electro-hyperthermia (mEHT), induced by 13.56 MHz radiofrequency, has been demonstrated both in preclinical and clinical studies to efficiently induce tumor damage and complement other treatment modalities. Here, we used a mouse xenograft model of human melanoma (A2058) to test mEHT (~42°C) both alone and combined with NK-cell immunotherapy. A single 30 min shot of mEHT resulted in significant tumor damage due to induced stress, marked by high hsp70 expression followed by significant upregulation of cleaved/activated caspase-3 and p53. When mEHT was combined with either primary human NK cells or the IL-2 independent NK-92MI cell line injected subcutaneously, the accumulation of NK cells was observed at the mEHT pretreated melanoma nodules but not at the untreated controls. mEHT induced the upregulation of the chemoattractant CXCL11 and increased the expression of the matrix metalloproteinase MMP2 which could account for the NK-cell attraction into the treated melanoma. In conclusion, mEHT monotherapy of melanoma xenograft tumors induced irreversible heat and cell stress leading to caspase dependent apoptosis to be driven by p53. mEHT could support the intratumoral attraction of distantly injected NK-cells, contributed by CXCL11 and MMP2 upregulation, resulting in an additive tumor destruction and growth inhibition. Therefore, mEHT may offer itself as a good partner for immunotherapy.
Collapse
Affiliation(s)
- Tamás Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | | | - Anett Benedek
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Ralf Bergmann
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
5
|
Zhang W, Zhu L, An C, Wang R, Yang L, Yu W, Li P, Gao Y. The blood brain barrier in cerebral ischemic injury – Disruption and repair. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2019.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
6
|
Modulation of in vitro Brain Endothelium by Mechanical Trauma: Structural and Functional Restoration by Poloxamer 188. Sci Rep 2020; 10:3054. [PMID: 32080247 PMCID: PMC7033190 DOI: 10.1038/s41598-020-59888-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 01/31/2020] [Indexed: 11/24/2022] Open
Abstract
Brain injuries caused by an explosive blast or blunt force is typically presumed to associate with mechanical trauma to the brain tissue. Recent findings from our laboratory suggest that shockwaves produced by a blast can generate micron-sized bubbles in the tissue. The collapse of microbubbles (i.e., microcavitation) may induce a mechanical trauma and compromise the integrity of the blood-brain endothelium (BBE). To test our hypothesis, we engineered a BBE model to determine the effect of microbubbles on the structural and functional changes in the BBE. Using monolayers of mouse primary brain microvascular endothelial cells, the permeability coefficient was measured following simulated blast-induced microcavitation. This event down-regulated the expression of tight junction markers, disorganized the cell-cell junction, and increased permeability. Since poloxamers have been shown to rescue damaged cells, the cells were treated with the FDA-approved poloxamer 188 (P188). The results indicate P188 recovered the permeability, restored the tight junctions, and suppressed the expressions of matrix metalloproteinases. The biomimetic interface we developed appears to provide a systematic approach to replicate the structure and function of BBE, determine its alteration in response to traumatic brain injury, and test potential therapeutic treatments to repair the damaged brain endothelium.
Collapse
|
7
|
Ruszkiewicz JA, Tinkov AA, Skalny AV, Siokas V, Dardiotis E, Tsatsakis A, Bowman AB, da Rocha JBT, Aschner M. Brain diseases in changing climate. ENVIRONMENTAL RESEARCH 2019; 177:108637. [PMID: 31416010 PMCID: PMC6717544 DOI: 10.1016/j.envres.2019.108637] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 05/12/2023]
Abstract
Climate change is one of the biggest and most urgent challenges for the 21st century. Rising average temperatures and ocean levels, altered precipitation patterns and increased occurrence of extreme weather events affect not only the global landscape and ecosystem, but also human health. Multiple environmental factors influence the onset and severity of human diseases and changing climate may have a great impact on these factors. Climate shifts disrupt the quantity and quality of water, increase environmental pollution, change the distribution of pathogens and severely impacts food production - all of which are important regarding public health. This paper focuses on brain health and provides an overview of climate change impacts on risk factors specific to brain diseases and disorders. We also discuss emerging hazards in brain health due to mitigation and adaptation strategies in response to climate changes.
Collapse
Affiliation(s)
- Joanna A Ruszkiewicz
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alexey A Tinkov
- Yaroslavl State University, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cellular and Intracellular Symbiosis, Russian Academy of Sciences, Orenburg, Russia
| | - Anatoly V Skalny
- Yaroslavl State University, Yaroslavl, Russia; IM Sechenov First Moscow State Medical University, Moscow, Russia; Trace Element Institute for UNESCO, Lyon, France
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, 71003, Heraklion, Greece
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, United States
| | - João B T da Rocha
- Department of Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
8
|
Sun M, Shinoda Y, Fukunaga K. KY-226 Protects Blood-brain Barrier Function Through the Akt/FoxO1 Signaling Pathway in Brain Ischemia. Neuroscience 2018; 399:89-102. [PMID: 30579831 DOI: 10.1016/j.neuroscience.2018.12.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/12/2018] [Accepted: 12/16/2018] [Indexed: 01/05/2023]
Abstract
KY-226 is a protein tyrosine phosphatase 1B (PTP1B) inhibitor that protects neurons from cerebral ischemic injury. KY-226 restores Akt (protein kinase B) phosphorylation and extracellular signal-regulated kinase (ERK) reduction in transient middle cerebral artery occlusion (tMCAO) damage. However, the mechanisms underlying the neuroprotective effects of KY-226 are unclear. To address this, the effects of KY-226 on blood-brain barrier (BBB) dysfunction were examined in tMCAO mice. KY-226 (10 mg/kg, i.p.) was administered to ICR mice 30 min after 2 h of tMCAO. To assess Akt or ERK involvement, wortmannin (i.c.v.) or U0126 (i.v.), selective inhibitors of PI3K and ERK, respectively, were administered to mice 30 min before ischemia. BBB integrity was assessed by Evans blue leakage 24 h post-reperfusion. The levels of tight junction (TJ) proteins, ZO-1 and occludin, were measured by western blotting; ZO-1 mRNA level was measured by RT-PCR. Compared to vehicle, KY-226 treatment prevented BBB breakdown and reduction in TJ protein levels. KY-226 treatment restored ZO-1 mRNA levels post-reperfusion. Pre-administration of wortmannin or U0126 blocked the protective effects of KY-226 on ZO-1 protein and mRNA reduction in tMCAO mice. In bEnd.3 cells, lipopolysaccharide treatment reduced mRNA and protein levels of ZO-1, an effect rescued by KY-226 treatment. Further, KY-226 treatment restored phosphorylation of pAkt (T308) and its downstream target forkhead box protein O1 (FoxO1) (S256) in bEnd.3 cells. Collectively, we demonstrate that KY-226 protects BBB integrity by restoration of TJ proteins, an effect partly mediated by Akt/FoxO1 pathway activation. Thus, protection of BBB integrity likely underlies KY-226-induced neuroprotection in tMCAO mice.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan.
| |
Collapse
|
9
|
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
|
10
|
Isaacson KJ, Martin Jensen M, Subrahmanyam NB, Ghandehari H. Matrix-metalloproteinases as targets for controlled delivery in cancer: An analysis of upregulation and expression. J Control Release 2017; 259:62-75. [PMID: 28153760 DOI: 10.1016/j.jconrel.2017.01.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/18/2017] [Accepted: 01/26/2017] [Indexed: 02/07/2023]
Abstract
While commonly known for degradation of the extracellular matrix, matrix metalloproteinases (MMPs) exhibit broad potential for use in targeting of bioactive and imaging agents in cancer treatment. MMPs are upregulated at all stages of expression in cancers. A comprehensive analysis of published literature on expression of all MMP subtypes at the genetic, protein, and activity levels in normal and diseased tissues indicate targeting applicability in a variety of cancers. This expression significantly increases at advanced cancer stages, providing an improved opportunity for controlled release in higher-stage patients. Since MMPs are integral at every stage of metastasis, MMP roles in cancer are discussed with a focus on MMP distribution and mobility within cells and tumors for cancer targeting applications. Several strategies for MMP utilization in targeting - such as matrix degradation, MMP cleavage, MMP binding, and MMP-induced environmental changes - are addressed.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Nithya B Subrahmanyam
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
11
|
Release of Matrix Metalloproteinases-2 and 9 by S-Nitrosylated Caveolin-1 Contributes to Degradation of Extracellular Matrix in tPA-Treated Hypoxic Endothelial Cells. PLoS One 2016; 11:e0149269. [PMID: 26881424 PMCID: PMC4755609 DOI: 10.1371/journal.pone.0149269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 01/30/2016] [Indexed: 11/19/2022] Open
Abstract
Intracranial hemorrhage remains the most feared complication in tissue plasminogen activator (tPA) thrombolysis for ischemic stroke. However, the underlying molecular mechanisms are still poorly elucidated. In this study, we reported an important role of caveolin-1 (Cav-1) s-nitrosylation in matrix metalloproteinase (MMP)-2 and 9 secretion from tPA-treated ischemic endothelial cells. Brain vascular endothelial cells (bEND3) were exposed to oxygen-glucose deprivation (OGD) for 2 h before adding recombinant human tPA for 6 h. This treatment induced a significant increase of MMP2 and 9 in the media of bEND3 cells and a simultaneous degradation of fibronectin and laminin β-1, the two main components of extracellular matrix (ECM). Inhibition of MMP2 and 9 with SB-3CT completely blocked the degradation of fibronectin and laminin β-1. ODG+tPA treatment led to Cav-1 shedding from bEND3 cells into the media. Notably, OGD triggered nitric oxide (NO) production and S-nitrosylationof Cav-1 (SNCav-1). Meanwhile tPA induced activation of ERK signal pathway and stimulates the secretion of SNCav-1. Pretreatment of bEND3 cells with C-PTIO (a NO scavenger) or U0126 (a specific ERK inhibitor) significantly reduced OGD-induced S-nitrosylation of Cav-1 in cells and blocked the secretion of Cav-1 and MMP2 and 9 into the media as well as the degradation of fibronectin and laminin β-1 in OGD and tPA-treated cells. These data indicate that OGD-triggered Cav-1 S-nitrosylation interacts with tPA-induced ERK activation to augment MMP2 and 9 secretion and subsequent ECM degradation, which may account for the exacerbation of ischemic blood brain barrier damage following tPA thrombolysis for ischemic stroke.
Collapse
|
12
|
Ji C, Wang L, Dai R, Shan L, Yang H, Zhu H, Meng Q. Hyperthermia exacerbates the effects of cathepsin L on claudin-1 in a blood-brain barrier model in vitro. Brain Res 2015; 1631:72-9. [PMID: 26655064 DOI: 10.1016/j.brainres.2015.11.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 01/17/2023]
Abstract
PURPOSE The effects of cathepsin L on claudin-1 expression were investigated under hyperthermic condition in a blood-brain barrier (BBB) model in vitro, in order to estimate the potential effects of hyperthermia on BBB dysfunction. MATERIALS AND METHODS Brain microvascular endothelial cells (BMECs) and astrocytes were obtained from rat brain. The BBB models were randomly divided into a sham (37°C) group, a 39°C group, a 37°C+cathepsin L group and a 39°C+cathepsin L group. The permeability of BBB was judged. The expressions of cathepsin L in astrocytes and claudin-1 in BMECs were detected using immunohistochemistry method and western blot assay. RESULTS The permeability of BBB models was higher in the 39°C group than in the sham group. The cathepsin L expression in astrocytes was higher in the 39°C group than in the sham group (P<0.01), whereas the claudin-1 expression in BMECs was lower in the 39°C group than in the sham group (P<0.01). The claudin-1 expression in BMECs was significantly lower in the 37°C+cathepsin L group than in the sham group (P<0.01). At the same time point, the claudin-1 expression in BMECs was significantly lower in the 39°C+cathepsin L group than in the 37°C+cathepsin L group (P<0.01). CONCLUSION Hyperthermia can probably decrease claudin-1 expression in BMECs by upregulating cathepsin L expression in astrocytes in a BBB model in vitro.
Collapse
Affiliation(s)
- Conghua Ji
- Department of Postgraduate, Kunming Medical University, Kunming, Yunnan, China
| | - Limin Wang
- Department of Postgraduate, Kunming Medical University, Kunming, Yunnan, China
| | - Rongrong Dai
- Department of Clinical Laboratory, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li Shan
- Department of Postgraduate, Kunming Medical University, Kunming, Yunnan, China
| | - Hui Yang
- Department of Pathology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Hongyan Zhu
- Department of Clinical Laboratory, the Affiliated Kunhua Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiang Meng
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| |
Collapse
|
13
|
Hou H, Zhang G, Wang H, Gong H, Wang C, Zhang X. High matrix metalloproteinase-9 expression induces angiogenesis and basement membrane degradation in stroke-prone spontaneously hypertensive rats after cerebral infarction. Neural Regen Res 2014; 9:1154-62. [PMID: 25206775 PMCID: PMC4146104 DOI: 10.4103/1673-5374.135318] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2014] [Indexed: 11/13/2022] Open
Abstract
Basement membrane degradation and blood-brain barrier damage appear after cerebral infarction, severely impacting neuronal and brain functioning; however, the underlying pathogenetic mechanisms remain poorly understood. In this study, we induced cerebral infarction in stroke-prone spontaneously hypertensive rats by intragastric administration of high-sodium water (1.3% NaCl) for 7 consecutive weeks. Immunohistochemical and immunofluorescence assays demonstrated that, compared with the non-infarcted contralateral hemisphere, stroke-prone spontaneously hypertensive rats on normal sodium intake and Wistar-Kyoto rats, matrix metalloproteinase-9 expression, the number of blood vessels with discontinuous collagen IV expression and microvessel density were significantly higher, and the number of continuous collagen IV-positive blood vessels was lower in the infarct border zones of stroke-prone spontaneously hypertensive rats given high-sodium water. Linear correlation analysis showed matrix metalloproteinase-9 expression was positively correlated with the number of discontinuously collagen IV-labeled blood vessels and microvessel density in cerebral infarcts of stroke-prone spontaneously hypertensive rats. These results suggest that matrix metalloproteinase-9 upregulation is associated with increased regional angiogenesis and degradation of collagen IV, the major component of the basal lamina, in stroke-prone spontaneously hypertensive rats with high-sodium water-induced focal cerebral infarction.
Collapse
Affiliation(s)
- Huilian Hou
- Department of Pathology, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Guanjun Zhang
- Department of Pathology, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hongyan Wang
- Department of Pathology, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Huilin Gong
- Department of Pathology, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chunbao Wang
- Department of Pathology, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xuebin Zhang
- Department of Pathology, the First Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
14
|
Wu L, Xu L, Xu X, Fan X, Xie Y, Yang L, Lan W, Zhu J, Xu G, Dai J, Jiang Y, Liu X. Keep warm and get success: The role of postischemic temperature in the mouse middle cerebral artery occlusion model. Brain Res Bull 2014; 101:12-7. [DOI: 10.1016/j.brainresbull.2013.12.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 11/27/2013] [Accepted: 12/04/2013] [Indexed: 12/14/2022]
|
15
|
Li DD, Song JN, Huang H, Guo XY, An JY, Zhang M, Li Y, Sun P, Pang HG, Zhao YL, Wang JF. The roles of MMP-9/TIMP-1 in cerebral edema following experimental acute cerebral infarction in rats. Neurosci Lett 2013; 550:168-72. [DOI: 10.1016/j.neulet.2013.06.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/04/2013] [Accepted: 06/17/2013] [Indexed: 11/27/2022]
|
16
|
Abstract
Ischaemic stroke is one of the leading causes of death and disability worldwide, and intravenous alteplase is the only proven effective treatment in the acute setting. Hypothermia has been shown to improve neurological outcomes after global ischaemia-hypoxia in comatose patients who have had cardiac arrest, and is one of the most extensively studied and powerful therapeutic strategies in acute ischaemic stroke. The protective mechanisms of therapeutic hypothermia affect the ischaemic cascade across several parallel pathways and, when coupled with reperfusion strategies, might yield synergistic benefits for patients who have had a stroke. Technological advances have allowed hypothermia to be induced rapidly, and the treatment has been used safely in acute stroke patients. Conclusive efficacy trials assessing therapeutic hypothermia combined with reperfusion therapies in acute ischaemic stroke are ongoing.
Collapse
|
17
|
Alam M, Shuaib A. Complexity in differentiating the expression of truncated or matured forms of MMP-2 and MMP-9 through zymography in rat brain tissues after acute ischaemic stroke. J Neurosci Methods 2013; 216:22-7. [PMID: 23523510 DOI: 10.1016/j.jneumeth.2013.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 03/12/2013] [Indexed: 10/27/2022]
Abstract
Matrix metalloproteinases (MMPs) play an important role in the pathogenesis of ischaemic stroke. In particular, the mature forms of MMPs 2 and 9 have similar sizes and share gelatine as a common substrate. Both MMPs are upregulated in ischaemic stroke and play detrimental roles during stroke pathogenesis. Throughout this study, we demonstrated that pro-MMP-2 and pro-MMP-9 from ischaemic rat brain tissue homogenate is detected either through immunoblotting or zymography because of the remarkable size difference between these enzymes (72 versus 95 kDa, respectively). However, the mature MMP-2 and MMP-9 cannot be discriminated through zymography because of the almost identical sizes of these forms (66 and 67 kDa, respectively). The use of gelatine zymography on ischaemic rat brain tissue homogenate revealed a 65-kDa MMP band, corresponding to the heterogeneous band of mature MMP-2 and/or MMP-9. Furthermore, we also detected mature MMPs of 65 kDa generated from both recombinant human MMP-2 and MMP-9. Using a pull down assay in rat brain tissue homogenate with gelatine-agarose beads, we showed increased activities for both the pro and mature forms of MMP-2 and MMP-9. However, we could not determine the origin of the respective mature MMPs from the heterogeneous band. Thus, in this study, we demonstrated that the identification and quantification of mature MMP-2 and MMP-9 could not be achieved using zymography alone. Therefore, the development of a reliable technique to identify and measure the respective MMPs is needed to test new stroke therapies targeting MMP-2 and MMP-9.
Collapse
Affiliation(s)
- Mustafa Alam
- Stroke Research Laboratory, Department of Medicine-Neurology, University of Alberta, Canada
| | | |
Collapse
|
18
|
Human matrix metalloproteinases: an ubiquitarian class of enzymes involved in several pathological processes. Mol Aspects Med 2011; 33:119-208. [PMID: 22100792 DOI: 10.1016/j.mam.2011.10.015] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 10/29/2011] [Indexed: 02/07/2023]
Abstract
Human matrix metalloproteinases (MMPs) belong to the M10 family of the MA clan of endopeptidases. They are ubiquitarian enzymes, structurally characterized by an active site where a Zn(2+) atom, coordinated by three histidines, plays the catalytic role, assisted by a glutamic acid as a general base. Various MMPs display different domain composition, which is very important for macromolecular substrates recognition. Substrate specificity is very different among MMPs, being often associated to their cellular compartmentalization and/or cellular type where they are expressed. An extensive review of the different MMPs structural and functional features is integrated with their pathological role in several types of diseases, spanning from cancer to cardiovascular diseases and to neurodegeneration. It emerges a very complex and crucial role played by these enzymes in many physiological and pathological processes.
Collapse
|