1.Evaluation of the diagnostic utility of urine biomarkers Tissue Inhibitor of Metalloproteinases-2 (TIMP-2) and Insulin-like Growth Factor Binding Protein-7 (IGFBP-7) in predicting acute kidney injury and short-term outcomes among high-risk, critically ill.
Renz Michael F. Pasilan ; Bab E. Pangan ; John Jefferson V. Besa ; Daniel Y. Guevara ; Jonnel B. Poblete ; Maria Charissa Thalia M. Pornillos ; Maria Isabel D. Duavit
Acta Medica Philippina 2024;58(16):14-22
BACKGROUND AND OBJECTIVES
Acute kidney injury (AKI) is a common complication of critical illness that often leads to increased mortality and morbidity. Biomarkers detect AKI earlier, providing a window of opportunity for timely intervention. Of the recent biomarkers in literature, the cell cycle arrest biomarkers tissue inhibitor of metalloproteinases-2 (TIMP-2) and insulin-like growth factor binding protein-7 (IGFBP-7) were found to be superior in predicting AKI. Our study aimed to evaluate the diagnostic performance of urine TIMP-2/IGFBP-7 in its ability to predict AKI and major adverse kidney events within 30 days (MAKE30) among high-risk patients for AKI. MAKE30 is a composite outcome comprised of all-cause mortality, use of renal replacement therapy (RRT), or persistent renal dysfunction at hospital discharge truncated at 30 days.
METHODSWe conducted a prospective, cross-sectional study which included 135 adult, non-COVID ICU patients. Baseline urine TIMP-2/IGFBP-7 results were used to dichotomize the population into low risk (< 0.3 ng/mL) or high risk (≥ 0.3 ng/mL) for AKI. Participants were then observed for 30 days and monitored for MAKE30 outcomes. ROC curves were created to calculate the sensitivity, specificity, NPV, PPV, and the AUC of the 0.3 ng/mL cut-off to predict the AKI and MAKE30.
RESULTSUrine TIMP-2/IGFBP-7 cutoff of 0.3 ng/mL predicted AKI with a sensitivity of 82.4%, specificity of 79.2%, PPV of 57.1%, NPV of 93% and AUC of 0.81. MAKE30 was detected with a sensitivity of 62.8%, specificity of 76.1%, PPV of 55.1%, NPV of 81.4% and AUC of 0.69. Elevated levels of urine TIMP-2/IGFBP-7 were found to be associated with AKI (p <0.01), MAKE30 (p <0.01) and all of its subcomponents. Survival or discharge after 30 days were found to be associated with lower urine TIMP-2/IGFBP-7 levels (p <0.01).
CONCLUSIONUrine TIMP-2/IGFBP-7, at its current cutoff at 0.3 ng/mL, can predict the likelihood of developing AKI and major adverse kidney events among high-risk patients for AKI. It can serve as a useful adjunct to existing methods, such as serum creatinine, in the early diagnosis and prognosis of acute kidney injury and expanding the therapeutic window to prevent disease progression and improve outcomes.
Acute Kidney Injury ; Biomarkers ; Urine ; Tissue Inhibitor Of Metalloproteinase-2 ; Insulin-like Growth Factor Binding Proteins
2.Overwork Affects Extracellular Matrix of Arterial Vessel Wall in Rats.
Su-Heng CHEN ; Lu GAN ; Miao ZHUANG ; Xiao-Xiao ZHANG ; Hong GUO ; Rong-Rong HUANG ; Yu-Lan LI
Acta Academiae Medicinae Sinicae 2022;44(2):262-269
Objective To explore the effect of overwork (OW) on extracellular matrix of arterial vessel wall in rats. Methods Random number grouping method was employed to assign 18 Sprague-Dawley rats into three groups(n=6):the control group(no special treatment),group OW(forced swimming twice a day for 15 days),and sleep deficiency(SD)+OW group(in addition to forced swimming twice a day,the rats were put on the platforms in water to limit sleep for 15 days).On the 16th day,the abdominal aorta and common carotid artery were collected after blood sampling from heart under deep anesthesia.A part of the abdominal aorta sample was taken for Masson staining of collagen fiber,and Verhoeff-Van Gieson staining was carried out for the elastic fiber of common carotid artery.Image J was employed for the quantitative analysis of collagen fiber and elastic fiber content.The expression of collagen 1(Col-1) protein was quantified by immunohistochemistry and the ultrastructure of vascular matrix was examined by transmission electron microscopy.The other part of the abdominal aorta sample was used to determine the mRNA levels of matrix metalloproteinase(MMP)-1,MMP-2,MMP-9,tissue inhibitor of metalloproteinases-1(TIMP-1),and Col-1 by quantitative real-time polymerase chain reaction. Results Compared with that in control group,the content of collagen fiber in groups OW and SD+OW had no significant change(all P>0.05);the content of elastic fiber in groups OW and SD+OW decreased(all P<0.001) and had no significant difference between each other(P>0.05).The vascular vessel wall of group OW showed slight fiber breakage,while that of group SD+OW presented wormhole-like or spongy fiber fragmentation.The mRNA levels of MMP-1 and MMP-2 in groups OW and SD+OW had no significant difference between each other(P>0.05) but were higher than that in control group(all P<0.001).The mRNA levels of MMP-9 and TIMP-1 had no significant difference among the three groups(all P>0.05).Groups OW and SD+OW had lower mRNA level(all P<0.001) and protein level(all P<0.001) of Col-1 than control group,while the mRNA and protein levels of Col-1 had no significant difference between groups OW and SD+OW(P>0.05). Conclusion OW can reduce the content of Col-1 and elastic fibers in the extracellular matrix of arterial vessels,destroy the elastic lamina of vascular wall,up-regulate the expression of MMP-1 and MMP-2,thereby injuring arterial vessels.
Animals
;
Collagen Type I
;
Extracellular Matrix/metabolism*
;
Matrix Metalloproteinase 1/metabolism*
;
Matrix Metalloproteinase 2/metabolism*
;
Matrix Metalloproteinase 9/metabolism*
;
RNA, Messenger/genetics*
;
Rats
;
Rats, Sprague-Dawley
;
Tissue Inhibitor of Metalloproteinase-1/metabolism*
3.The myocardial protective effect of propofol on rats with experimental myocardial infarction and its mechanism.
Ming-Xiao ZHANG ; Qing-Xin TIAN ; Jian-Long LIU
Acta Physiologica Sinica 2021;73(6):878-884
The aim of the present study was to investigate the protective effect of propofol on the experimental myocardial infarction in rats. The myocardial infarction model was established by ligating the anterior descending branch of left coronary artery in rats. Model rats were treated with propofol. Cardiac function was evaluated by echocardiography. Cardiac hemodynamic changes were detected by multiconductor biorecorder. Pathological changes in the infarcted myocardia were detected by HE staining. The expression levels of cardiac hypertrophy marker genes and fibrosis marker proteins were analyzed by real-time quantitative PCR and Western blot. The results showed that, compared with the sham surgery group, the model group exhibited larger infarct size (> 40%), impaired heart function, and significantly increased left ventricular end-diastolic pressure (LVEDP). Propofol reduced cardiac function impairment and decreased LVEDP in the model group. Propofol significantly reduced lung weight/body weight ratio, heart weight/body weight ratio, left ventricular weight/body weight ratio and left atrial weight/body weight ratio in the model group. Furthermore, after myocardial infarction, the administration of propofol significantly improved the diastolic strain rate, down-regulated the mRNA expression levels of myocardial hypertrophy markers, atrial natriuretic peptide and β-myosin heavy chain, and reversed the up-regulation of matrix metalloproteinase 2 (MMP2), MMP9 and tissue inhibitor of metalloproteinase-2 (TIMP-2) induced by myocardial infarction. These results suggest propofol can reduce adverse ventricular remodeling, cardiac dysfunction, myocardial hypertrophy and fibrosis after myocardial infarction, and has protective effect against the experimental myocardial infarction induced by coronary artery ligation in rats.
Animals
;
Cardiotonic Agents/pharmacology*
;
Matrix Metalloproteinase 2
;
Matrix Metalloproteinase 9
;
Myocardial Infarction/drug therapy*
;
Myocardium
;
Propofol/pharmacology*
;
Rats
;
Tissue Inhibitor of Metalloproteinase-2/genetics*
;
Ventricular Remodeling
4.Prokaryotic expression, purification and characterization of tissue inhibitor of metalloproteinase-2.
Aiying XUE ; Guoxing FENG ; Changchun ZHU ; Saijun FAN
Chinese Journal of Biotechnology 2020;36(12):2868-2876
Tissue inhibitor of metalloproteinases-2 (TIMP-2) inhibits tumor migration and invasion. Obtaining TIMP-2 protein is conducive to a comprehensive and in-depth study of its function and mechanism in tumorigenesis and development. We collected human TIMP-2 protein through prokaryotic expression in vitro. We expressed, purified and characterized human TIMP-2 protein. First, the human TIMP-2 gene was cloned from the cDNA obtained by reverse transcription of total RNA of human lung cancer A549 cells, and constructed to pET28a vector. The recombinant plasmid pET28a-TIMP-2 was transformed into Escherichia coli BL21(DE3) after restriction endonuclease digestion and sequencing analysis. The expression of TIMP-2 protein was induced by isopropyl-β-D-thiogalactoside (IPTG), and the expression conditions were optimized. After purification by nickel affinity column, the fusion protein His-TIMP-2 was identified by Western blotting method and its biological activity was detected by gelatin zymography. The fusion protein His-TIMP-2 existed in the form of inclusion body in E. coli. In a certain range, the concentration of IPTG had no significant effect on the expression amount of His-TIMP-2. But in this expression system, induction temperature and time were the key parameters, and the expression amount of His-TIMP-2 in E. coli increased with the increase of induction temperature. The purified and refolded fusion protein could effectively inhibit the activity of matrix metalloproteinases expressed by human lung cancer A549 cells. The acquisition of active fusion protein lays a foundation for further study of the function and mechanism of human TIMP-2, and is of great significance for tumor therapy.
Cloning, Molecular
;
Escherichia coli/genetics*
;
Humans
;
Recombinant Fusion Proteins/genetics*
;
Recombinant Proteins
;
Tissue Inhibitor of Metalloproteinase-2/genetics*
5.Enhanced Anti-Cancer Effects of Conditioned Medium from Hypoxic Human Umbilical Cord–Derived Mesenchymal Stem Cells
Kyu Hyun HAN ; Ae Kyeong KIM ; Gun Jae JEONG ; Hye Ran JEON ; Suk Ho BHANG ; Dong Ik KIM
International Journal of Stem Cells 2019;12(2):291-303
BACKGROUND AND OBJECTIVES: There have been contradictory reports on the pro-cancer or anti-cancer effects of mesenchymal stem cells. In this study, we investigated whether conditioned medium (CM) from hypoxic human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) (H-CM) showed enhanced anti-cancer effects compared with CM from normoxic hUC-MSCs (N-CM). METHODS AND RESULTS: Compared with N-CM, H-CM not only strongly reduced cell viability and increased apoptosis of human cervical cancer cells (HeLa cells), but also increased caspase-3/7 activity, decreased mitochondrial membrane potential (MMP), and induced cell cycle arrest. In contrast, cell viability, apoptosis, MMP, and cell cycle of human dermal fibroblast (hDFs) were not significantly changed by either CM whereas caspase-3/7 activity was decreased by H-CM. Protein antibody array showed that activin A, Beta IG-H3, TIMP-2, RET, and IGFBP-3 were upregulated in H-CM compared with N-CM. Intracellular proteins that were upregulated by H-CM in HeLa cells were represented by apoptosis and cell cycle arrest terms of biological processes of Gene Ontology (GO), and by cell cycle of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. In hDFs, negative regulation of apoptosis in biological process of GO and PI3K-Akt signaling pathway of KEGG pathways were represented. CONCLUSIONS: H-CM showed enhanced anti-cancer effects on HeLa cells but did not influence cell viability or apoptosis of hDFs and these different effects were supported by profiling of secretory proteins in both kinds of CM and intracellular signaling of HeLa cells and hDFs.
Activins
;
Anoxia
;
Apoptosis
;
Biological Processes
;
Cell Cycle
;
Cell Cycle Checkpoints
;
Cell Survival
;
Culture Media, Conditioned
;
Fibroblasts
;
Gene Ontology
;
Genome
;
HeLa Cells
;
Humans
;
Insulin-Like Growth Factor Binding Protein 3
;
Membrane Potential, Mitochondrial
;
Mesenchymal Stromal Cells
;
Tissue Inhibitor of Metalloproteinase-2
;
Uterine Cervical Neoplasms
6.Coexistence of proangiogenic potential and increased MMP-9, TIMP-1, and TIMP-2 levels in the plasma of patients with critical limb ischemia.
Radosław WIECZÓR ; Anna Maria WIECZÓR ; Arleta KULWAS ; Grzegorz PULKOWSKI ; Jacek BUDZYŃSKI ; Danuta ROŚĆ
Journal of Zhejiang University. Science. B 2019;20(8):687-692
The objective of this study was to assess the angiogenic potential expressed as a quotient of vascular endothelial growth factor A (VEGF-A), as an indicator of proangiogenic activity, and the circulating receptors (soluble VEGF receptor protein R1 (sVEGFR-1) and sVEGFR-2), as indicators of the effect of angiogenic inhibition, depending on the concentrations of matrix metalloproteinase 2 (MMP-2) and MMP-9 and their tissue inhibitor 1 (TIMP-1) and TIMP-2 in the plasma of patients with lower extremity artery disease (LEAD). These blood parameters in patients with intermittent claudication (IC) and critical limb ischemia (CLI) were compared for select clinical and biochemical features. Stimulation of angiogenesis in the plasma of individuals with LEAD was evident as indicated by the significant increase in VEGF-A concentration along with reduced inhibition depending on circulating receptors sVEGFR-1 and sVEGFR-2. Critical ischemia was associated with higher VEGF-A, MMP-9, TIMP-1, and TIMP-2 concentrations than in the case of IC.
Aged
;
Angiogenesis Inhibitors/pharmacology*
;
Female
;
Gene Expression Regulation
;
Humans
;
Intermittent Claudication/drug therapy*
;
Ischemia/drug therapy*
;
Lower Extremity/blood supply*
;
Male
;
Matrix Metalloproteinase 9/blood*
;
Middle Aged
;
Neovascularization, Pathologic
;
Tissue Inhibitor of Metalloproteinase-1/blood*
;
Tissue Inhibitor of Metalloproteinase-2/blood*
;
Vascular Endothelial Growth Factor A/blood*
;
Vascular Endothelial Growth Factor Receptor-1/blood*
;
Vascular Endothelial Growth Factor Receptor-2/blood*
7.Regulation of Matrix Metalloproteinase 2 Expression by an Adenosine A1 Agonist in Trabecular Meshwork Cells
Min Ju BAEK ; Keun Hae KIM ; Jae Woo KIM
Journal of the Korean Ophthalmological Society 2018;59(10):946-952
PURPOSE: We investigated the extent of adenosine A1 agonist-induced expression and regulation of matrix metalloproteinase 2 (MMP-2) synthesis in human trabecular meshwork cells (HTMC). METHODS: Primary HTMC cultures were exposed to 0.1 or 1.0 µM N6-cyclohexyladenosine (CHA) for 2 h in the presence or absence of an inhibitor thereof, 8-cyclopentyl-1,3-dimethylxanthine (CPT). The expression level of mRNA encoding MMP-2 was assessed via reverse transcription-polymerase chain reaction, and the levels of tissue inhibitor of metalloproteinase 2 (TIMP2) and membrane-type-1 MMP (MT1-MMP) measured by Western blotting. The permeability of the HTMC monolayer was assessed with the aid of carboxyfluorescein. RESULTS: CHA at 1.0 µM increased the permeability of the HTMC monolayer (p = 0.003) and CHA at both 0.1 and 1.0 µM significantly increased MMP-2 mRNA expression, which was inhibited by co-exposure to CPT (all p < 0.05). CHA increased MMP-2 activity, decreased that of TIMP2, and increased that of MT1-MMP (all p < 0.05). CONCLUSIONS: CHA increased the permeability of the HTMC monolayer and increased MMP-2 activity, decreased TIMP2 activity, and increased MT1-MMP activity. Thus, regulation of TIMP2 and MT1-MMP expression may be involved in the adenosine A1 agonist-induced increase in MMP-2 activity.
Adenosine
;
Blotting, Western
;
Humans
;
Matrix Metalloproteinase 14
;
Matrix Metalloproteinase 2
;
Permeability
;
RNA, Messenger
;
Tissue Inhibitor of Metalloproteinase-2
;
Trabecular Meshwork
8.Diagnostic Value of Sensitive Biomarkers for Early Kidney Damage in Diabetic Patients with Normoalbuminuria.
Dong ZHANG ; Qiu-Xia HAN ; Ming-Hui WU ; Wan-Jun SHEN ; Xiao-Li YANG ; Jia GUO ; Shao-Kang PAN ; Zhang-Suo LIU ; Li TANG ; Guang-Yan CAI ; Xiang-Mei CHEN ; Han-Yu ZHU
Chinese Medical Journal 2018;131(23):2891-2892
Biomarkers
;
urine
;
Cross-Sectional Studies
;
Diabetes Mellitus, Type 2
;
pathology
;
urine
;
Enzyme-Linked Immunosorbent Assay
;
Hepatitis A Virus Cellular Receptor 1
;
metabolism
;
Humans
;
Intracellular Signaling Peptides and Proteins
;
urine
;
Kidney Diseases
;
pathology
;
urine
;
Lipocalin-2
;
urine
;
Membrane Proteins
;
urine
;
Sialoglycoproteins
;
urine
;
Tissue Inhibitor of Metalloproteinase-2
;
urine
9.Effect of subcutaneous treatment with human umbilical cord blood-derived multipotent stem cells on peripheral neuropathic pain in rats.
Min Ju LEE ; Tae Gyoon YOON ; Moonkyu KANG ; Hyun Jeong KIM ; Kyung Sun KANG
The Korean Journal of Physiology and Pharmacology 2017;21(2):153-160
In this study, we aim to determine the in vivo effect of human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs) on neuropathic pain, using three, principal peripheral neuropathic pain models. Four weeks after hUCB-MSC transplantation, we observed significant antinociceptive effect in hUCB-MSC–transplanted rats compared to that in the vehicle-treated control. Spinal cord cells positive for c-fos, CGRP, p-ERK, p-p 38, MMP-9 and MMP 2 were significantly decreased in only CCI model of hUCB-MSCs-grafted rats, while spinal cord cells positive for CGRP, p-ERK and MMP-2 significantly decreased in SNL model of hUCB-MSCs-grafted rats and spinal cord cells positive for CGRP and MMP-2 significantly decreased in SNI model of hUCB-MSCs-grafted rats, compared to the control 4 weeks or 8weeks after transplantation (p<0.05). However, cells positive for TIMP-2, an endogenous tissue inhibitor of MMP-2, were significantly increased in SNL and SNI models of hUCB-MSCs-grafted rats. Taken together, subcutaneous injection of hUCB-MSCs may have an antinociceptive effect via modulation of pain signaling during pain signal processing within the nervous system, especially for CCI model. Thus, subcutaneous administration of hUCB-MSCs might be beneficial for improving those patients suffering from neuropathic pain by decreasing neuropathic pain activation factors, while increasing neuropathic pain inhibition factor.
Animals
;
Cord Blood Stem Cell Transplantation
;
Humans*
;
Injections, Subcutaneous
;
Multipotent Stem Cells*
;
Nervous System
;
Neuralgia*
;
Rats*
;
Spinal Cord
;
Tissue Inhibitor of Metalloproteinase-2
;
Umbilical Cord*
10.Resveratrol raises in vitro anticancer effects of paclitaxel in NSCLC cell line A549 through COX-2 expression.
Fanhua KONG ; Runqi ZHANG ; Xudong ZHAO ; Guanlin ZHENG ; Zhou WANG ; Peng WANG
The Korean Journal of Physiology and Pharmacology 2017;21(5):465-474
The aim of this study was to determine the raising anticancer effects of resveratrol (Res) on paclitaxel (PA) in non-small cell lung cancer (NSCLC) cell line A549. The 10 µg/ml of Res had no effect on human fetal lung fibroblast MRC-5 cells or on A549 cancer cells and the 5 or 10 µg/ml of PA also had no effect on MRC-5 normal cells. PA-L (5 µg/ml) and PA-H (10 µg/ml) had the growth inhibitory effects in NSCLC cell line A549, and Res increased these growth inhibitory effects. By flow cytometry experiment, after Res (5 µg/ml)+PA-H (10 µg/ml) treatment, the A549 cells showed the most apoptosic cells compared to other group treatments, and after additional treatment with Res, the apoptosic cells of both two PA concentrations were raised. Res+PA could reduce the mRNA and protein expressions of COX-2, and Res+PA could reduce the COX-2 related genes of VEGF, MMP-1, MMP-2, MMP-9, NF-κB, Bcl-2, Bcl-xL, procollagen I, collagen I, collagen III and CTGF, TNF-α, IL-1β, iNOS and raise the TIMP-1, TIMP-2, TIMP-3, IκB-α, p53, p21, caspase-3, caspase-8, caspase-9, Bax genes compared to the control cells and the PA treated cells. From these results, it can be suggested that Res could raise the anticancer effects of PA in A549 cells, thus Res might be used as a good sensitizing agent for PA.
Carcinoma, Non-Small-Cell Lung
;
Caspase 3
;
Caspase 8
;
Caspase 9
;
Cell Line*
;
Collagen
;
Fibroblasts
;
Flow Cytometry
;
Humans
;
In Vitro Techniques*
;
Lung
;
Paclitaxel*
;
Procollagen
;
RNA, Messenger
;
Tissue Inhibitor of Metalloproteinase-1
;
Tissue Inhibitor of Metalloproteinase-2
;
Tissue Inhibitor of Metalloproteinase-3
;
Vascular Endothelial Growth Factor A


Result Analysis
Print
Save
E-mail