Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 264
Filter
1.
Mol Cancer ; 23(1): 70, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38576002

ABSTRACT

BACKGROUND: Cellular senescence frequently occurs during anti-cancer treatment, and persistent senescent tumor cells (STCs) unfavorably promote tumor progression through paracrine secretion of the senescence-associated secretory phenotype (SASP). Extracellular vesicles (EVs) have recently emerged as a novel component of the SASP and primarily mediate the tumor-promoting effect of the SASP. Of note, the potential effect of EVs released from STCs on tumor progression remains largely unknown. METHODS: We collected tumor tissues from two cohorts of colorectal cancer (CRC) patients to examine the expression of p16, p21, and SERPINE1 before and after anti-cancer treatment. Cohort 1 included 22 patients with locally advanced rectal cancer (LARC) who received neoadjuvant therapy before surgical resection. Cohort 2 included 30 patients with metastatic CRC (mCRC) who received first-line irinotecan-contained treatment. CCK-8, transwell, wound-healing assay, and tumor xenograft experiments were carried out to determine the impacts of EVs released from STCs on CRC progression in vitro and in vivo. Quantitative proteomic analysis was applied to identify protein cargo inside EVs secreted from STCs. Immunoprecipitation and mass spectrometer identification were utilized to explore the binding partners of SERPINE1. The interaction of SERPINE1 with p65 was verified by co-immunoprecipitation, and their co-localization was confirmed by immunofluorescence. RESULTS: Chemotherapeutic agents and irradiation could potently induce senescence in CRC cells in vitro and in human CRC tissues. The more significant elevation of p16 and p21 expression in patients after anti-cancer treatment displayed shorter disease-free survival (DFS) for LARC or progression-free survival (PFS) for mCRC. We observed that compared to non-STCs, STCs released an increased number of EVs enriched in SERPINE1, which further promoted the progression of recipient cancer cells. Targeting SERPINE1 with a specific inhibitor, tiplaxtinin, markedly attenuated the tumor-promoting effect of STCs-derived EVs. Additionally, the patients with greater increment of SERPINE1 expression after anti-cancer treatment had shorter DFS for LARC or PFS for mCRC. Mechanistically, SERPINE1 bound to p65, promoting its nuclear translocation and subsequently activating the NF-κB signaling pathway. CONCLUSIONS: We provide the in vivo evidence of the clinical prognostic implications of therapy-induced senescence. Our results revealed that STCs were responsible for CRC progression by producing large amounts of EVs enriched in SERPINE1. These findings further confirm the crucial role of therapy-induced senescence in tumor progression and offer a potential therapeutic strategy for CRC treatment.


Subject(s)
Colorectal Neoplasms , Extracellular Vesicles , Rectal Neoplasms , Humans , NF-kappa B/metabolism , Proteomics , Signal Transduction , Extracellular Vesicles/metabolism , Rectal Neoplasms/metabolism , Cellular Senescence , Colorectal Neoplasms/pathology , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology
2.
J Thromb Thrombolysis ; 57(4): 721-729, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38523179

ABSTRACT

Hypercoagulability and reduced fibrinolysis are well-established complications associated with COVID-19. However, the timelines for the onset and resolution of these complications remain unclear. The aim of this study was to evaluate, in a cohort of COVID-19 patients, changes in coagulation and fibrinolytic activity through ROTEM assay at different time points during the initial 30 days following the onset of symptoms in both mild and severe cases. Blood samples were collected at five intervals after symptoms onset: 6-10 days, 11-15 days, 16-20 days, 21-25 days, and 26-30 days. In addition, fibrinogen, plasminogen, PAI-1, and alpha 2-antiplasmin activities were determined. Out of 85 participants, 71% had mild COVID-19. Twenty uninfected individuals were evaluated as controls. ROTEM parameters showed a hypercoagulable state among mild COVID-19 patients beginning in the second week of symptoms onset, with a trend towards reversal after the third week of symptoms. In severe COVID-19 cases, hypercoagulability was observed since the first few days of symptoms, with a tendency towards reversal after the fourth week of symptoms onset. A hypofibrinolytic state was identified in severe COVID-19 patients from early stages and persisted even after 30 days of symptoms. Elevated activity of PAI-1 and alpha 2-antiplasmin was also detected in severe COVID-19 patients. In conclusion, both mild and severe cases of COVID-19 exhibited transient hypercoagulability, reverted by the end of the first month. However, severe COVID-19 cases sustain hypofibrinolysis throughout the course of the disease, which is associated with elevated activity of fibrinolysis inhibitors. Persistent hypofibrinolysis could contribute to long COVID-19 manifestations.


Subject(s)
Antifibrinolytic Agents , COVID-19 , Thrombophilia , Humans , Fibrinolysis , Plasminogen Activator Inhibitor 1/pharmacology , Post-Acute COVID-19 Syndrome
3.
Diabetes Metab J ; 48(1): 72-82, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38173367

ABSTRACT

BACKGRUOUND: Renal fibrosis is characterized by the accumulation of extracellular matrix proteins and interstitial fibrosis. Alantolactone is known to exert anticancer, anti-inflammatory, antimicrobial and antifungal effects; however, its effects on renal fibrosis remains unknown. Here, we investigated whether alantolactone attenuates renal fibrosis in mice unilateral ureteral obstruction (UUO) and evaluated the effect of alantolactone on transforming growth factor (TGF) signaling pathway in renal cells. METHODS: To evaluate the therapeutic effect of alantolactone, cell counting kit-8 (CCK-8) assay, histological staining, Western blot analysis, and real-time quantitative polymerase chain reaction were performed in UUO kidneys in vivo and in TGF-ß-treated renal cells in vitro. RESULTS: Alantolactone (0.25 to 4 µM) did not affect the viability of renal cells. Mice orally administered 5 mg/kg of alantolactone daily for 15 days did not show mortality or liver toxicity. Alantolactone decreased UUO-induced blood urea nitrogen and serum creatinine levels. In addition, it significantly alleviated renal tubulointerstitial damage and fibrosis and decreased collagen type I, fibronectin, and α-smooth muscle actin (α-SMA) expression in UUO kidneys. In NRK-49F cells, alantolactone inhibited TGF-ßstimulated expression of fibronectin, collagen type I, plasminogen activator inhibitor-1 (PAI-1), and α-SMA. In HK-2 cells, alantolactone inhibited TGF-ß-stimulated expression of collagen type I and PAI-1. Alantolactone inhibited UUO-induced phosphorylation of Smad3 in UUO kidneys. In addition, it not only decreased TGF-ß secretion but also Smad3 phosphorylation and translocation to nucleus in both kidney cell lines. CONCLUSION: Alantolactone improves renal fibrosis by inhibiting the TGF-ß/Smad3 signaling pathway in obstructive nephropathy. Thus, alantolactone is a potential therapeutic agent for chronic kidney disease.


Subject(s)
Kidney Diseases , Lactones , Sesquiterpenes, Eudesmane , Ureteral Obstruction , Mice , Animals , Fibronectins/pharmacology , Fibronectins/therapeutic use , Plasminogen Activator Inhibitor 1/pharmacology , Plasminogen Activator Inhibitor 1/therapeutic use , Collagen Type I/pharmacology , Collagen Type I/therapeutic use , Kidney Diseases/drug therapy , Kidney Diseases/etiology , Ureteral Obstruction/complications , Ureteral Obstruction/drug therapy , Ureteral Obstruction/metabolism , Transforming Growth Factor beta/metabolism , Signal Transduction , Fibrosis
4.
Kidney Int ; 104(3): 508-525, 2023 09.
Article in English | MEDLINE | ID: mdl-37356621

ABSTRACT

Natriuretic peptides exert not only blood-lowering but also kidney-protective effects through guanylyl cyclase-A (GC-A), a natriuretic peptide receptor. Signaling through GC-A has been shown to protect podocytes from aldosterone-induced glomerular injury, and a p38 mitogen-activated protein kinase (MAPK) inhibitor reduced glomerular injury in aldosterone-infused podocyte-specific GC-A knockout mice. To explore the role of p38 MAPK in podocytes, we constructed podocyte-specific p38 MAPK and GC-A double knockout mice (pod-double knockout mice). Unexpectedly, aldosterone-infused and high salt-fed (B-ALDO)-treated pod-double knockout mice resulted in elevated serum creatinine, massive albuminuria, macrophage infiltration, foot process effacement, nephrin and podocin reduction, and additionally, intra-capillary fibrin thrombi, indicating endothelial injury. Microarray analysis showed increased plasminogen activator inhibitor-1 (PAI-1) in glomeruli of B-ALDO-treated pod-double knockout mice. In B-ALDO-treated pod-double knockout mice, PAI-1 increased in podocytes, and treatment with PAI-1 neutralizing antibody ameliorated intra-capillary thrombus formation. In vitro, deletion of p38 MAPK by the CRISPR/Cas9 system and knockdown of GC-A in human cultured podocytes upregulated PAI-1 and transforming growth factor- ß1 (TGF-ß1). When p38 MAPK knockout podocytes, transfected with a small interfering RNA to suppress GC-A, were co-cultured with glomerular endothelial cells in a transwell system, the expression of TGF-ß1 was increased in glomerular endothelial cells. PAI-1 inhibition ameliorated both podocyte and endothelial injury in the transwell system signifying elevated PAI-1 in podocytes is a factor disrupting normal podocyte-endothelial crosstalk. Thus, our results indicate that genetic dual deletion of p38 MAPK and GC-A in podocytes accelerates both podocyte and endothelial injuries, suggesting these two molecules play indispensable roles in podocyte function.


Subject(s)
Podocytes , Thrombosis , Animals , Humans , Mice , Aldosterone/pharmacology , Aldosterone/metabolism , Endothelial Cells/metabolism , Guanylate Cyclase/metabolism , Guanylate Cyclase/pharmacology , Mice, Knockout , p38 Mitogen-Activated Protein Kinases/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Podocytes/metabolism , Thrombosis/metabolism , Transforming Growth Factor beta1/metabolism , Mitogen-Activated Protein Kinase 14
5.
Int J Radiat Biol ; 99(5): 791-806, 2023.
Article in English | MEDLINE | ID: mdl-36383216

ABSTRACT

PURPOSE: Atherosclerosis is a lipid-driven chronic inflammatory disease that causes cardiovascular diseases (CVD). The association between radiation and atherosclerosis has already been demonstrated; however, the effects of low-dose radiation (LDR) exposure on atherosclerosis have not been reported. Our study aims to propose that LDR may cause atherosclerosis phenotypes by the upregulation of plasminogen activator inhibitor-1 (PAI-1) and downregulation of androgen receptor (AR), which are cytokines secreted from the liver. METHODS: Low-density lipoprotein (LDL) receptor deficient (Ldlr-/-) mice were irradiated at 50 mGy, 100 mGy, and 1000 mGy. LDR irradiated Ldlr-/- mice serum was analyzed by cytokine array and proteomics with silver staining. Oil Red O staining and BODIPY staining were performed to determine lipid accumulation in Human umbilical vein endothelial cells (HUVECs). Foam cell formation and monocyte recruitment were assessed through co-culture system with HUVECs and THP-1 cells. RESULTS: After irradiation with LDR (100 mGy) the mice showed atherosclerotic phenotypes and through analysis results, we selected regulated cytokines, PAI-1 and AR, and found that these were changed in the liver. LDR-regulated cytokines have the potential to be transported to endothelial cells and induce lipid accumulation, inflammation of monocytes, increased oxidized low-density lipoprotein (oxLDL) and foam cells formation, that were series of phenotypes lead to plaque formation in endothelial cells and induces atherosclerosis. As a further aspect of this study, testosterone undecanoate (TU) was found to pharmacologically inhibit a series of atherosclerotic phenotypes exhibited by LDR. This study suggests a role for PAI-1 and AR in regulating the development of atherosclerosis after LDR exposure. Targeting PAI-1 and AR could serve as an attractive strategy for the management of atherosclerosis following LDR exposure.


Subject(s)
Atherosclerosis , Cytokines , Humans , Animals , Mice , Plasminogen Activator Inhibitor 1/pharmacology , Atherosclerosis/etiology , Lipoproteins, LDL/pharmacology , Human Umbilical Vein Endothelial Cells , Liver
6.
Ther Hypothermia Temp Manag ; 13(3): 112-119, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36473198

ABSTRACT

Hypothermia affects coagulation, but the effect of hypothermia on fibrinolysis is not clarified. Imbalance in the fibrinolytic system may lead to increased risk of bleeding or thrombosis. Our aim was to investigate if resuscitated cardiac arrest patients treated with hypothermia had an unbalanced fibrinolysis. A prospective cohort study, including 82 patients were treated with hypothermia at 33°C ± 1°C after out-of-hospital cardiac arrest. Blood samples were collected at 24 hours (hypothermia) and at 72 hours (normothermia). Samples were analyzed for fibrin D-dimer, tissue plasminogen activator (tPA), plasminogen, plasminogen activator Inhibitor-1 (PAI-1), thrombin-activatable fibrinolysis inhibitor (TAFI), and an in-house dynamic fibrin clot formation and lysis assay.Compared with normothermia, hypothermia significantly increased plasminogen activity (mean difference = 10.4%, 95% confidence interval [CI] 7.9-12.9), p < 0.001), PAI-1 levels (mean difference = 275 ng/mL, 95% CI 203-348, p < 0.001), and tPA levels (mean difference = 1.0 ng/mL, 95% CI 0.2-1.7, p = 0.01). No differences between hypothermia and normothermia were found in TAFI activity (p = 0.59) or in the fibrin D-dimer levels (p = 0.08). The fibrin clot lysis curves showed three different patterns: normal-, flat-, or resistant clot lysis curve. At hypothermia 45 (55%) patients had a resistant clot lysis curve and 33 (44%) patients had a resistant clot lysis curve at normothermia (p = 0.047). Comatose, resuscitated, cardiac arrest patients treated with hypothermia express an inhibited fibrinolysis even after rewarming. This could potentially increase the thromboembolic risk. ClinicalTrials.gov ID: NCT02258360.


Subject(s)
Hypothermia, Induced , Hypothermia , Out-of-Hospital Cardiac Arrest , Humans , Fibrinolysis , Tissue Plasminogen Activator/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Prospective Studies , Hypothermia, Induced/adverse effects , Fibrin/pharmacology , Plasminogen/pharmacology , Out-of-Hospital Cardiac Arrest/therapy
7.
Nutr J ; 21(1): 65, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36258233

ABSTRACT

BACKGROUND: It is known that consuming a high-fat meal (HFM) induces microvascular dysfunction (MD) in eutrophic women and aggravates it in those with obesity. Our purpose was to investigate if the MD observed after a single HFM intake is caused by endothelial damage or increased inflammatory state, both determined by blood biomarkers. METHODS: Nineteen women with obesity (BMI 30-34.9 kg/m2) and 18 eutrophic ones (BMI 20.0-24.9 kg/m2) were enrolled into two groups: Obese (OBG) and Control (CG), respectively. Blood samples were collected at five-time points: before (fasting state) and 30, 60, 120, and 180 min after HFM intake to determine levels of adipokines (adiponectin, leptin), non-esterified fatty acid (NEFA), inflammatory [tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6)] and endothelium damage [soluble E-selectin, soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular adhesion molecule-1 (sICAM-1), plasminogen activator inhibitor-1 (PAI-1)] biomarkers. RESULTS: Levels of soluble E-selectin, leptin, and PAI-1 were higher in OBG at all-time points (P < 0.05) compared to CG. In the fasting state, OBG had higher levels of NEFA compared to CG (P < 0.05). In intra-group analysis, no significant change in the levels of circulating inflammatory and endothelial injury biomarkers was observed after HFM intake, independently of the group. CONCLUSION: Our findings suggest that women with obesity have an increased pro-inflammatory state and more significant endothelial injury compared to eutrophic ones. However, the consumption of a HFM was not sufficient to change circulating levels of inflammatory and endothelial injury biomarkers in either group. REGISTRATION NUMBER FOR CLINICAL TRIALS: NCT01692327.


Subject(s)
Adiposity , Leptin , Female , Humans , Adipokines , Adiponectin , Biomarkers , Cross-Sectional Studies , E-Selectin/metabolism , Endothelium, Vascular , Fatty Acids, Nonesterified , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/pharmacology , Interleukin-6 , Obesity , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Tumor Necrosis Factor-alpha , Vascular Cell Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/pharmacology
8.
J Adv Res ; 40: 95-107, 2022 09.
Article in English | MEDLINE | ID: mdl-36100336

ABSTRACT

INTRODUCTION: Basic fibroblast growth factor (bFGF) plays a critical role in odontoblast differentiation and dentin matrix deposition, thereby aiding pulpo-dentin repair and regeneration. OBJECTIVES: The purpose of this study was to clarify the effects of bFGF on plasminogen activation factors, TIMP-1), ALP; and SPARC (osteonectin) expression/production of stem cells from apical papilla (SCAP) in vitro; and the involvement of MEK/ERK, p38, Akt, and TAK1 signaling. METHODS: SCAP were exposed to bFGF with/without pretreatment and co-incubation with various signal transduction inhibitors (U0126, SB203580, LY294002, and 5Z-7-oxozeaenol). The expression of FGF receptors (FGFRs), PAI-1, uPA, p-ERK, p-TAK1, and p-p38 was analyzed via immunofluorescent staining. The gene expression and protein secretion of SCAP were determined via real-time PCR and ELISA. ALP activity was evaluated via ALP staining. RESULTS: SCAP expressed FGFR1, 2, 3, and 4. bFGF stimulated the PAI-1, uPA, uPAR, and TIMP-1 mRNA expression (p < 0.05). bFGF induced PAI-1, uPA, and soluble uPAR production (p < 0.05) but suppressed the ALP activity and SPARC production (p < 0.05) of SCAP. bFGF stimulated ERK, TAK1, and p38 phosphorylation of SCAP. U0126 (a MEK/ERK inhibitor) and 5Z-7-oxozeaenol (a TAK1 inhibitor) attenuated the bFGF-induced PAI-1, uPA, uPAR, and TIMP-1 expression and production of SCAP, but SB203580 (a p38 inhibitor) did not. LY294002, SB203580, and 5Z-7oxozeaenol could not reverse the inhibition of ALP activity caused by bFGF. Interestingly, U0126 and 5Z-7-oxozeaenol prevented the bFGF-induced decline of SPARC production (p < 0.05). CONCLUSION: bFGF may regulate fibrinolysis and matrix turnover via modulation of PAI-1, uPA, uPAR, and TIMP-1, but bFGF inhibited the differentiation (ALP, SPARC) of SCAP. These events are mainly regulated by MEK/ERK, p38, and TAK1. Combined use of bFGF and SCAP may facilitate pulpal/root repair and regeneration via regulation of the plasminogen activation system, migration, matrix turnover, and differentiation of SCAP.


Subject(s)
Alkaline Phosphatase , Fibroblast Growth Factor 2 , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/pharmacology , Butadienes , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Lactones , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/pharmacology , Nitriles , Osteonectin/metabolism , Osteonectin/pharmacology , Plasminogen/metabolism , Plasminogen/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Resorcinols , Signal Transduction , Stem Cells/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Zearalenone/administration & dosage
9.
Arch Oral Biol ; 141: 105486, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35779427

ABSTRACT

OBJECTIVES: The aims of this study were to investigate the efficacy of Histatin-1 in wound closure as well as effects on gene expression of nicotine-treated human Periodontal Ligament Fibroblast cells (HPDL) in vitro. DESIGN: HPDL grown in 2.5% culture medium treated with 10 ng/ml Histatin - 1 in the presence/absence of 0.5 µM nicotine were subjected to wound assay and migration was studied at 0 h, 6 h, 12 h and 24 h. Cells grown in 2.5% medium served as control. Cell migration was studied by wound gap and transwell migration assays. The effect of Histatin-1 on expression of matrix metalloproteinase 8 (MMP-8), insulin-like growth factor 1 (IGF-1), transforming growth factor beta (TGF-ß), collagen type I (COL1) and plasminogen activator inhibitor 1 (PAI-1) were studied. RESULTS: Histatin-1 treatment significantly decreased percentage wound gap at 12 h (62.96 ± 3.22 vs 79.23 ± 1.73; p < 0.05) and at 24 h (38.78 ± 7.59 vs 75.21 ± 4.94; p < 0.001) compared with controls. In nicotine+Histatin-1 treated cells, wound gap decreased to 70.2 ± 2.9% (p < 0.01) at 24 h compared to nicotine alone in which 82 ± 1.64% of wound gap was retained. Transwell migration assays showed significant migration of HPDL with Histatin-1 (p < 0.05). Gene expression demonstrated significant upregulation for IGF-1, TGF ß, COL1 and PAI-1 with Histatin-1. CONCLUSION: Histatin-1 significantly mitigated the effect of nicotine in wound healing assay involving HPDL fibroblast cells at 24 h. Histatin-1 aided wound closure is attributed to the upregulation of IGF-1, TGF ß, COL1, and PAI-1 genes.


Subject(s)
Nicotine , Periodontal Ligament , Cells, Cultured , Fibroblasts , Histatins/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Nicotine/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Salivary Proteins and Peptides/metabolism , Transforming Growth Factor beta/metabolism
10.
J Appl Toxicol ; 42(5): 738-749, 2022 05.
Article in English | MEDLINE | ID: mdl-34708887

ABSTRACT

Fine particulate matter (PM2.5 )-induced detrimental cardiovascular effects have been widely concerned, especially for endothelial cells, which is the first barrier of the cardiovascular system. Among potential mechanisms involved, reactive oxidative species take up a crucial part. However, source of oxidative stress and its relationship with inflammatory response have been rarely studied in PM2.5 -induced endothelial injury. Here, as a key oxidase that catalyzes redox reactions, NADPH oxidase (NOX) was investigated. Human umbilical vein endothelial cells (EA.hy926) were exposed to Standard Reference Material 1648a of urban PM2.5 for 24 h, which resulted in NOX-sourced oxidative stress, endothelial dysfunction, and inflammation induction. These are manifested by the up-regulation of NOX, increase of superoxide anion and hydrogen peroxide, elevated endothelin-1 (ET-1) and asymmetric dimethylarginine (ADMA) level, reduced nitric oxide (NO) production, and down-regulation of phosphorylation of endothelial NO synthase (eNOS) with increased levels of inducible NO synthase, as well as the imbalance between tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor 1 (PAI-1), and changes in the levels of pro-inflammatory and anti-inflammatory factors. However, administration of NOX1/4 inhibitor GKT137831 alleviated PM2.5 -induced elevated endothelial dysfunction biomarkers (NO, ET-1, ADMA, iNOS, and tPA/PAI-1), inflammatory factors (IL-1ß, IL-10, and IL-18), and adhesion molecules (ICAM-1, VCAM-1, and P-selectin) and also passivated NOX-dependent AKT and eNOS phosphorylation that involved in endothelial activation. In summary, PM2.5 -induced NOX up-regulation is the source of ROS in EA.hy926, which activated AKT/eNOS/NO signal response leading to endothelial dysfunction and inflammatory damage in EA.hy926 cells.


Subject(s)
NADPH Oxidases , Nitric Oxide , Human Umbilical Vein Endothelial Cells , Humans , Particulate Matter/toxicity , Plasminogen Activator Inhibitor 1/pharmacology , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species
11.
Neurologist ; 27(6): 299-303, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-34855657

ABSTRACT

BACKGROUND: To: (i) explore the effect of diterpene ginkgolides meglumine injection (DGMI) on neurological deficit symptoms in acute atherosclerotic cerebral infarction (AACI) patients; (ii) measure the level of plasma plasminogen activator inhibitor (PAI)-1 and tissue plasminogen activator (t-PA). METHODS: Eighty AACI patients were divided equally and randomly into the DGMI group and control group. In addition to basic treatment, the DGMI group was treated with DGMI (25 mg/d) for 14 days. The control group had basic treatment without DGMI. Before and after treatment, the degree of neurological deficit was assessed, thromboelastography undertaken, and plasma levels of PAI-1 and t-PA measured. RESULTS: The National Institutes of Health Stroke Scale score of patients in the DGMI group after treatment was lower than that in the control group, and the Barthel Index was higher than that in the control group ( P <0.05). Thromboelastography revealed that, in the DGMI group, the R value and K value after treatment were higher than before treatment, the angle and maximum amplitude value were lower than before treatment, and both were significant ( P <0.05). Compared with the control group, the plasma PAI-1 level of patients in the DGMI group was lower than that in the control group, and the t-PA level was higher than that in the control group ( P <0.05) after 14 days of treatment. CONCLUSIONS: DGMI may affect the activity of the blood coagulation and fibrinolysis system by regulating the plasma level of PAI-1 and t-PA, and improving neurological deficit symptoms. DGMI is important for improving the prognosis of patients with AACI.


Subject(s)
Brain Ischemia , Stroke , Humans , Tissue Plasminogen Activator/therapeutic use , Tissue Plasminogen Activator/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Ginkgolides/pharmacology , Ginkgolides/therapeutic use , Fibrinolysis , Meglumine/pharmacology , Acute Disease , Cerebral Infarction/drug therapy
12.
Environ Pollut ; 287: 117283, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34426376

ABSTRACT

Numerous studies have established that acute or chronic exposure to environmental pollutants like particulate matter (PM) leads to the development of accelerated aging related pathologies including pulmonary and cardiovascular diseases, and thus air pollution is one of the major global threats to human health. Air pollutant particulate matter 2.5 (PM2.5)-induced cellular dysfunction impairs tissue homeostasis and causes vascular and cardiopulmonary damage. To test a hypothesis that elevated plasminogen activator inhibitor-1 (PAI-1) levels play a pivotal role in air pollutant-induced cardiopulmonary pathologies, we examined the efficacy of a drug-like novel inhibitor of PAI-1, TM5614, in treating PM2.5-induced vascular and cardiopulmonary pathologies. Results from biochemical, histological, and immunohistochemical studies revealed that PM2.5 increases the circulating levels of PAI-1 and thrombin and that TM5614 treatment completely abrogates these effects in plasma. PM2.5 significantly augments the levels of pro-inflammatory cytokine interleukin-6 (IL-6) in bronchoalveolar lavage fluid (BALF), and this also can be reversed by TM5614, indicating its efficacy in amelioration of PM2.5-induced increases in inflammatory and pro-thrombotic factors. TM5614 reduces PM2.5-induced increased levels of inflammatory markers cluster of differentiation 107 b (Mac3) and phospho-signal transducer and activator of transcription-3 (pSTAT3), adhesion molecule vascular cell adhesion molecule 1 (VCAM1), and apoptotic marker cleaved caspase 3. Longer exposure to PM2.5 induces pulmonary and cardiac thrombosis, but TM5614 significantly ameliorates PM2.5-induced vascular thrombosis. TM5614 also reduces PM2.5-induced increased blood pressure and heart weight. In vitro cell culture studies revealed that PM2.5 induces the levels of PAI-1, type I collagen, fibronectin (Millipore), and sterol regulatory element binding protein-1 and 2 (SREBP-1 and SREBP-2), transcription factors that mediate profibrogenic signaling, in cardiac fibroblasts. TM5614 abrogated that stimulation, indicating that it may block PM2.5-induced PAI-1 and profibrogenic signaling through suppression of SREBP-1 and 2. Furthermore, TM5614 blocked PM2.5-mediated suppression of nuclear factor erythroid related factor 2 (Nrf2), a major antioxidant regulator, in cardiac fibroblasts. Pharmacological inhibition of PAI-1 with TM5614 is a promising therapeutic approach to control air pollutant PM2.5-induced cardiopulmonary and vascular pathologies.


Subject(s)
Air Pollutants , Air Pollution , Air Pollutants/toxicity , Humans , Lung , Particulate Matter/toxicity , Plasminogen Activator Inhibitor 1/pharmacology
13.
J Invest Dermatol ; 141(11): 2690-2698.e6, 2021 11.
Article in English | MEDLINE | ID: mdl-34000287

ABSTRACT

Immune checkpoint molecules, especially PD-1 and its ligand PD-L1, act as a major mechanism of cancer immune evasion. Although anti-PD-1/PD-L1 monotherapy increases therapeutic efficacy in melanoma treatment, only a subset of patients exhibits long-term tumor remission, and the underlying mechanism of resistance to PD-1/PD-L1 inhibitors remains unclear. In this study, we demonstrated that cell surface retention of PD-L1 is inversely correlated with PAI-1 expression in vitro, in vivo, and in clinical specimens. Moreover, extracellular PAI-1 induced the internalization of surface-expressed PD-L1 by triggering clathrin-mediated endocytosis. The endocytosed PD-L1 was transported to lysosomes for degradation by endolysosomal systems, resulting in the reduction of surface PD-L1. Notably, inhibition of PAI-1 by pharmacological inhibitor with tiplaxtinin led to elevated PD-L1 expression on the plasma membrane, both in vitro and in vivo. Strikingly, targeting PAI-1 by tiplaxtinin treatment synergizes with anti-PD-L1 immune checkpoint blockade therapy in a syngeneic murine model of melanoma. Our findings demonstrate a role for PAI-1 activity in immune checkpoint modulation by promoting surface PD-L1 for lysosomal degradation and provides an insight into the combination of PAI-1 inhibition and anti-PD-L1 immunotherapy as a promising therapeutic regimen for melanoma treatment.


Subject(s)
B7-H1 Antigen/metabolism , Endocytosis/drug effects , Immune Checkpoint Inhibitors/therapeutic use , Indoleacetic Acids/pharmacology , Melanoma/drug therapy , Plasminogen Activator Inhibitor 1/pharmacology , Animals , B7-H1 Antigen/analysis , B7-H1 Antigen/antagonists & inhibitors , Caveolins/physiology , Humans , Indoleacetic Acids/therapeutic use , Melanoma/metabolism , Mice , Mice, Inbred C57BL
14.
Biochem Biophys Res Commun ; 557: 180-186, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33866038

ABSTRACT

Bone marrow adipocytes (BMAs) have recently been recognized as a niche component with a suppressive function. Obese individuals with abundant BMAs exhibit impaired hematopoietic regeneration after hematopoietic stem cell transplantation (HSCT). We hypothesized that plasminogen activator inhibitor type-1 (PAI-1), an adipokine that regulates the fibrinolytic system, contributes to impaired hematopoiesis in bone marrow (BM) microenvironment with abundant BMAs. We demonstrated that BMAs differentiated in vitro could secrete PAI-1 and were positive for PAI-1 in vivo. In addition, the abundance of BMAs was associated with high levels of PAI-1 expression. The BMA-rich microenvironment exhibited impaired hematopoietic regeneration after HSCT when compared with a BMA-less microenvironment. The impaired hematopoietic regeneration in BMA-rich microenvironment was significantly alleviated by PAI-1 knockout or PAI-1 inhibitor treatment. Obese mice with abundant BMAs, compared with normal-weight mice, exhibited higher bone marrow PAI-1 concentrations, increased fibrinolytic system suppression, and lower stem cell factor (SCF) concentrations after HSCT. PAI-1 inhibitor administration significantly activated the fibrinolytic system in obese mice, contributing to the higher SCF concentration. Moreover, PAI-1 inhibitor treatment significantly alleviated the impaired hematopoietic regeneration in obese mice both after 5-fluorouracil injection and HSCT. These results indicate that PAI-1 hinders hematopoietic regeneration in BMA-rich microenvironments. The blockade of PAI-1 activity could be a novel therapeutic means of facilitating hematopoietic reconstitution in BMA-rich patients.


Subject(s)
Adipocytes/metabolism , Bone Marrow/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Obesity/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Animals , Antimetabolites/pharmacology , Bone Marrow/metabolism , Fluorouracil/pharmacology , Gene Knockout Techniques , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/therapy , Plasminogen Activator Inhibitor 1/genetics , Regeneration/drug effects , Stem Cell Factor/metabolism , Stem Cell Niche/drug effects
15.
Elife ; 102021 04 27.
Article in English | MEDLINE | ID: mdl-33904394

ABSTRACT

Cortical interneurons establish inhibitory microcircuits throughout the neocortex and their dysfunction has been implicated in epilepsy and neuropsychiatric diseases. Developmentally, interneurons migrate from a distal progenitor domain in order to populate the neocortex - a process that occurs at a slower rate in humans than in mice. In this study, we sought to identify factors that regulate the rate of interneuron maturation across the two species. Using embryonic mouse development as a model system, we found that the process of initiating interneuron migration is regulated by blood vessels of the medial ganglionic eminence (MGE), an interneuron progenitor domain. We identified two endothelial cell-derived paracrine factors, SPARC and SerpinE1, that enhance interneuron migration in mouse MGE explants and organotypic cultures. Moreover, pre-treatment of human stem cell-derived interneurons (hSC-interneurons) with SPARC and SerpinE1 prior to transplantation into neonatal mouse cortex enhanced their migration and morphological elaboration in the host cortex. Further, SPARC and SerpinE1-treated hSC-interneurons also exhibited more mature electrophysiological characteristics compared to controls. Overall, our studies suggest a critical role for CNS vasculature in regulating interneuron developmental maturation in both mice and humans.


Subject(s)
Cell Movement/drug effects , Cerebral Cortex/metabolism , Induced Pluripotent Stem Cells/drug effects , Interneurons/drug effects , Median Eminence/blood supply , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Osteonectin/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Action Potentials , Animals , Cerebral Cortex/embryology , Cerebral Cortex/surgery , Endothelial Cells/metabolism , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Interneurons/metabolism , Interneurons/transplantation , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Median Eminence/embryology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Neovascularization, Physiologic , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Osteonectin/metabolism , Paracrine Communication , Plasminogen Activator Inhibitor 1/metabolism , Signal Transduction
16.
Lab Invest ; 101(3): 353-368, 2021 03.
Article in English | MEDLINE | ID: mdl-33311557

ABSTRACT

Cancer-associated fibroblasts (CAFs) contribute to the progression of various cancers. Previously, we reported the significance of CAFs in esophageal squamous cell carcinoma (ESCC); however, the functions of CAFs in the ESCC microenvironment remain unknown. To investigate CAFs' function, we established an indirect coculture assay between human bone marrow-derived mesenchymal stem cells (MSCs) and ESCC cells. Cocultured MSCs expressed more fibroblast activation protein, one of the markers of CAFs, compared with monocultured MSCs. Therefore, we defined cocultured MSCs as CAF-like cells. To identify molecules associated with the ESCC progression in CAFs, we conducted a cDNA microarray analysis on monocultured MSCs and CAF-like cells to compare their gene expression profiles. We found that SERPINE1, which encodes plasminogen activator inhibitor-1 (PAI-1), was more abundant in CAF-like cells than in monocultured MSCs, and the PAI-1 derived from CAF-like cells induced the abilities of migration and invasion in both ESCC cells and macrophages by the Akt and Erk1/2 signaling pathways via the low-density lipoprotein receptor-related protein 1 (LRP1), which is a PAI-1 receptor. Based on immunohistochemistry assays of ESCC tissues, higher expression levels of PAI-1 and LRP1 were correlated with poor prognosis in ESCC patients. These results suggest that the PAI-1/LRP1 axis contributes to the progression of ESCC, making it a potential target for ESCC therapy.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cell Movement/drug effects , Esophageal Squamous Cell Carcinoma/metabolism , Macrophages/drug effects , Plasminogen Activator Inhibitor 1/pharmacology , Aged , Cell Line, Tumor , Cells, Cultured , Esophageal Neoplasms/metabolism , Female , Humans , Macrophages/physiology , Male , Middle Aged , Neoplasm Invasiveness , Plasminogen Activator Inhibitor 1/metabolism
17.
IEEE Trans Nanobioscience ; 19(3): 446-456, 2020 07.
Article in English | MEDLINE | ID: mdl-32603295

ABSTRACT

AIM OF THE STUDY: In vitro evaluation of the effects of plasminogen activator inhibitor-1 (PAI-1) transfected-conditioned media (P-CM) on the differentiation of human periodontal ligament stem cells (hPDLSCs) and human periapical follicular stem cells (hPAFSCs). MATERIALS AND METHODS: The hPDLSCs and hPAFSCs received from impacted third molars were treated with P-CM and viability, as well as differentiation of the cells were evaluated. Plasmids were constructed according to standard techniques, and all sequences were validated by proper enzyme digestion and sequencing. Chinese hamster ovarian (CHO) cells were transfected with pcDNA3.1-hPAI-1 plasmid to obtain P-CM, followed by western blotting and PAI-1-specific ELISA kit to evaluate the proteins of P-CM. The cell viability of hPDLSCs and hPAFSCs were analyzed using MTT assay after 48 h of incubation. Alizarin red S staining was performed to evaluate the differentiation of hPDLSCs and hPAFSCs. The reverse transcription-polymerase chain reaction was used to observe the expression levels of osteogenic/cementogenic marker genes. The human cytokine antibody array was applied for further analysis of cytokine expression in P-CM. RESULTS: P-CM significantly promoted the differentiation of hPDLSCs and hPAFSCs and upregulated the expression of osteogenic/cementogenic marker genes in vitro. Furthermore, rhPAI-1 promoted mineralized nodules formation of hPDLSCs and hPAFSCs, and we identified that other proteins, RANTES and IL-6, were highly expressed in P-CM. CONCLUSIONS: P-CM promoted the differentiation of hPDLSCs and hPAFSCs by upregulating the expression of RANTES and IL-6, and interaction between PAI-1 and RANTES/IL-6 signaling may be involved in P-CM-induced osteogenic/cementogenic differentiation.


Subject(s)
Osteogenesis , Plasminogen Activator Inhibitor 1 , Stem Cells/metabolism , Transfection/methods , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Cytokines/metabolism , Humans , Osteogenesis/drug effects , Osteogenesis/genetics , Periapical Tissue/cytology , Periodontal Ligament/cytology , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
18.
Exp Cell Res ; 391(1): 111987, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32240661

ABSTRACT

BACKGROUND: The protein plasminogen activator inhibitor-1 (PAI-1), an inhibitor specific for urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA), has been shown to have a key role in cancer metastases. Currently, it is unknown as to whether the exocellular inhibition of PAI-1 can inhibit the migration of cancer cells. METHODS: By fusing the mutated serine protease domain (SPD) of uPA and human serum albumin (HSA), PAItrap3, a protein that traps PAI-1, was synthesized and experiments were conducted to determine if exocellular PAItrap3 attenuates PAI-1-induced cancer cell migration in vitro. RESULTS: PAItrap3 (0.8 µM) significantly inhibited the motility of MCF-7, MDA-MB-231, HeLa and 4T1 cancer cells, by 90%, 50%, 30% and 20%, respectively, without significantly altering their proliferation. The PAI-1-induced rearrangement of F-actin was significantly inhibited by PAItrap3, which produced a decrease in the number of cell protrusions by at least 20%. CONCLUSIONS: In vitro, PAItrap3 inhibited PAI-1-induced cancer cell migration, mainly through inhibiting the rearrangement of F-actin. Overall, these results, provided they can be extrapolated to humans, suggest that the PAItrap3 protein could be used as an exocellular inhibitor to attenuate cancer metastases.


Subject(s)
Actins/genetics , Cell Movement/drug effects , Plasminogen Activator Inhibitor 1/pharmacology , Protein C Inhibitor/pharmacology , Actins/antagonists & inhibitors , Actins/metabolism , Binding Sites , Cell Line , Cell Movement/genetics , Cell Proliferation/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Histidine/genetics , Histidine/metabolism , Humans , MCF-7 Cells , Oligopeptides/genetics , Oligopeptides/metabolism , Pichia/genetics , Pichia/metabolism , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Protein C Inhibitor/chemistry , Protein C Inhibitor/genetics , Protein C Inhibitor/metabolism , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
Cell Death Dis ; 10(10): 757, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31591377

ABSTRACT

PAI-1 plays significant roles in cancer occurrence, relapse and multidrug resistance and is highly expressed in tumours. ACT001, which is currently in phase I clinical trials for the treatment of glioblastoma (GBM). However, the detailed molecular mechanism of ACT001 is still unclear. In this study, we investigated the effects of ACT001 on glioma cell proliferation and clarified its mechanism. We discovered that PAI-1 was the direct target of ACT001 by a cellular thermal shift assay. Then, the interaction between ACT001 and PAI-1 was verified by Biacore assays, thermal stability assays and ACT001 probe assays. Furthermore, from the proteomic analysis, we found that ACT001 directly binds PAI-1 to inhibit the PI3K/AKT pathway, which induces the inhibition of glioma cell proliferation, invasion and migration. Moreover, the combination of ACT001 and cisplatin showed a synergistic effect on the inhibition of glioma in vitro and in vivo. In conclusion, our findings demonstrate that PAI-1 is a new target of ACT001, the inhibition of PAI-1 induces glioma inhibition, and ACT001 has a synergistic effect with cisplatin through the inhibition of the PAI-1/PI3K/AKT pathway.


Subject(s)
Cisplatin/pharmacology , Glioma/drug therapy , Plasminogen Activator Inhibitor 1/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Glioma/genetics , Glioma/pathology , Humans , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Phosphatidylinositol 3-Kinases/genetics , Plasminogen Activator Inhibitor 1/genetics , Proteomics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Reprod Domest Anim ; 54 Suppl 4: 65-68, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31625245

ABSTRACT

The serine proteases, tissue- and urokinase-type plasminogen activators (PLAT and PLAU) and their inhibitors SERPINE1/2 are regulators of plasminogen to plasmin conversion. They are widely expressed in ovarian tissues, including granulosa and cumulus cells, and their expression is regulated by gonadotropins. The aim of this work was to assess the effect of serine protease inhibitors (aprotinin and AEBSF) and SERPINE1/2 on FSH-induced cumulus cell expansion, the production of prostaglandin E2 (PGE2) and retention of hyaluronic acid (HA) in expanding cumulus. The serine protease activity proved to be essential for the production of PGE2 and also for the retention of HA; the inhibition of plasminogen activators by SERPINE1/2 had the same effect. Collectively, these data indicate that plasmin is required for proper function of expanding cumulus cells in vitro and presumably also in vivo in the pre-ovulatory follicles.


Subject(s)
Cumulus Cells/drug effects , Dinoprostone/metabolism , Oocytes/drug effects , Plasminogen Activator Inhibitor 1/pharmacology , Serine Proteinase Inhibitors/pharmacology , Serpin E2/pharmacology , Animals , Aprotinin/pharmacology , Cumulus Cells/cytology , Cumulus Cells/metabolism , Female , Follicle Stimulating Hormone/pharmacology , Hyaluronic Acid/metabolism , Oocytes/cytology , Oocytes/metabolism , Sulfones/pharmacology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...