Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 816
Filter
1.
PLoS One ; 17(2): e0263478, 2022.
Article in English | MEDLINE | ID: mdl-35113965

ABSTRACT

BACKGROUND: Autism spectrum disorder (ASD) is characterized by impaired social communication and behavioral problems. An increased risk of premature mortality has been observed in individuals with ASD. Therefore, we hypothesized that biological aging is accelerated in individuals with ASD. Recently, several studies have established genome-wide DNA methylation (DNAm) profiles as 'epigenetic clocks' that can estimate biological aging. In addition, ASD has been associated with differential DNAm patterns. METHODS: We used two independent datasets from blood samples consisting of adult patients with high-functioning ASD and controls: the 1st cohort (38 ASD cases and 31 controls) and the 2nd cohort (6 ASD cases and 10 controls). We explored well-studied epigenetic clocks such as HorvathAge, HannumAge, SkinBloodAge, PhenoAge, GrimAge, and DNAm-based telomere length (DNAmTL). In addition, we investigated seven DNAm-based age-related plasma proteins, including plasminogen activator inhibitor-1 (PAI-1), and smoking status, which are the components of GrimAge. RESULTS: Compared to controls, individuals with ASD in the 1st cohort, but not in the 2nd cohort, exhibited a trend for increased GrimAge acceleration and a significant increase of PAI-1 levels. A meta-analysis showed significantly increased PAI-1 levels in individuals with ASD compared to controls. CONCLUSION: Our findings suggest there is no epigenetic age acceleration in the blood of individuals with ASD. However, this study provides novel evidence regarding increased plasma PAI-1 levels in individuals with high-functioning ASD. These findings suggest PAI-1 may be a biomarker for high-functioning ASD, however, larger studies based on epigenetic clocks and PAI-1 will be necessary to confirm these findings.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Epigenesis, Genetic , Plasminogen Activator Inhibitor 1/biosynthesis , Adult , Biomarkers , Case-Control Studies , DNA Methylation , Female , Humans , Male , Regression Analysis , Risk , Telomere/ultrastructure , Young Adult
2.
Cells ; 10(1)2021 01 15.
Article in English | MEDLINE | ID: mdl-33467682

ABSTRACT

Abdominal aortic aneurysm (AAA) bears a high risk of rupture and sudden death of the patient. The pathogenic mechanisms of AAA remain elusive, and surgical intervention represents the only treatment option. Heme oxygenase-1 (HO-1), a heme degrading enzyme, is induced in AAA, both in mice and humans. HO-1 was reported to mitigate AAA development in an angiotensin II (AngII)-induced model of AAA in hyperlipidemic ApoE-/- mice. Since the role of hyperlipidaemia in the pathogenesis of AAA remains controversial, we aimed to evaluate the significance of HO-1 in the development and progression of AAA in normolipidemic animals. The experiments were performed in HO-1-deficient mice and their wild-type counterparts. We demonstrated in non-hypercholesterolemic mice that the high-dose of AngII leads to the efficient formation of AAA, which is attenuated by HO-1 deficiency. Yet, if formed, they are significantly more prone to rupture upon HO-1 shortage. Differential susceptibility to AAA formation does not rely on enhanced inflammatory response or oxidative stress. AAA-resistant mice are characterized by an increase in regulators of aortic remodeling and angiotensin receptor-2 expression, significant medial thickening, and delayed blood pressure elevation in response to AngII. To conclude, we unveil a dual role of HO-1 deficiency in AAA in normolipidemic mice, where it protects against AAA development, but exacerbates the state of formed AAA.


Subject(s)
Angiotensin II/adverse effects , Aortic Aneurysm, Abdominal/metabolism , Heme Oxygenase-1/metabolism , Membrane Proteins/metabolism , Oxidative Stress , Aneurysm/metabolism , Animals , Cardiovascular Diseases/metabolism , Cell Line , Collagen/metabolism , Genotype , Humans , Hyperlipidemias/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Receptor, Angiotensin, Type 2/metabolism , Serpin E2/metabolism , Skin/metabolism , Swine , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/metabolism
3.
Biomed Res Int ; 2020: 2406159, 2020.
Article in English | MEDLINE | ID: mdl-32104682

ABSTRACT

ELK3, an ETS domain-containing transcription factor, participates in various physiological and pathological processes including cell proliferation, migration, angiogenesis, and malignant progression. However, the role of ELK3 in prostate cancer cells and its mechanism are not fully understood. The contribution of ELK3 to prostate cancer progression was investigated in the present study. We showed that silencing of ELK3 by siRNA in prostate cancer cell DU145 induced S-M phase arrest, promoted apoptosis, inhibited cell proliferation and migration in vitro, and suppressed xenograft growth in mice in vivo. In accordance with its ability to arrest cells in S-M phase, the expression of cyclin A and cyclin B was downregulated. In addition, the expression of p53 was upregulated following ELK3 knockdown, while that of antiapoptotic Bcl-2 was decreased. The migration inhibition may partly due to upregulation of SERPINE1 (a serine protease inhibitor) followed ELK3 knockdown. Consistently, downregulation of SERPINE1 resulted in a modest elimination of migration inhibition resulted from ELK3 knockdown. Furthermore, we found that the AKT signaling was activated in ELK3 knockdown cells, and treatment these cells with AKT inhibitor attenuated SERPINE1 expression induced by ELK3 silencing, suggesting that activation of AKT pathway may be one of the reasons for upregulation of SERPINE1 after ELK3 knockdown. In conclusion, modulation of ELK3 expression may control the progression of prostate cancer partly by regulating cell growth, apoptosis, and migration.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Gene Silencing , M Phase Cell Cycle Checkpoints , Plasminogen Activator Inhibitor 1 , Prostatic Neoplasms , Proto-Oncogene Proteins c-ets , S Phase Cell Cycle Checkpoints , Up-Regulation , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/biosynthesis , Proto-Oncogene Proteins c-ets/genetics
4.
Exp Neurol ; 326: 113177, 2020 04.
Article in English | MEDLINE | ID: mdl-31926166

ABSTRACT

Preconditioning peripheral nerve injury primes the sensory neurons in the dorsal root ganglia (DRGs) to acquire axon regeneration competence. Transcription of a large set of regeneration-associated-genes (RAGs) contributes to the enhanced intrinsic axonal regeneration capacity. However, the mechanism underlying the coordinated upregulation of RAGs orchestrated by preconditioning injury is unclear. We sought to determine potential influence of DNA methylation change on transcriptional activation of RAGs in the L4-L6 DRGs following sciatic nerve injury. Genome-wide sequencing revealed that about 20% of the methylated DNA fragments were differentially methylated, and >3000 genes contained differentially methylated regions. Not only demethylation but also increased methylation was observed to a similar extent. The change in the global DNA methylation did not correlate with the gene expression level of most genes, including the well-documented RAGs. However, pharmacological inhibition or activation of DNA methylation markedly attenuated the axon growth capacity of the preconditioned DRG neurons. Pharmacological perturbation of DNA methylation resulted in simultaneous downregulation of many highly overlapping non-transcription factor RAGs, which was accompanied by a concurrent, robust upregulation of SOCS3 and Serpine1. Overexpression of SOCS3 and Serpine1 in the DRG neurons overrode injury-induced axon growth competence, corroborating their roles as the negative regulators of axon regeneration. We conclude that the injury-induced global alteration of DNA methylome strongly influences the axon growth competence in preconditioned DRG neurons. Our results also suggest a possibility that perturbing DNA methylome changes might lead to the upregulation of negative regulator RAGs thereby attenuating axon growth capacity.


Subject(s)
Axons/pathology , DNA Methylation , Ischemic Preconditioning , Peripheral Nerve Injuries/pathology , Sensory Receptor Cells/pathology , Animals , Cells, Cultured , DNA Methylation/drug effects , Ganglia, Spinal/cytology , Ganglia, Spinal/pathology , Gene Expression Regulation/genetics , Male , Nerve Regeneration/genetics , Neurites/drug effects , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Rats , Rats, Sprague-Dawley , Suppressor of Cytokine Signaling 3 Protein/biosynthesis , Suppressor of Cytokine Signaling 3 Protein/genetics , Transcriptional Activation
5.
Cancer Biomark ; 27(3): 285-293, 2020.
Article in English | MEDLINE | ID: mdl-31640087

ABSTRACT

OBJECTIVES: Thus far, biological roles of plasminogen activator inhibitor 1 (PAI1) in hepatocellular carcinoma (HCC) remain controversial. Moreover, its expression, clinicopathologic and prognostic significance in HCC have not been comprehensively investigated, therefore needing further evidence. METHODS: PAI1 expression was measured, using tissue microarray-based immunohistochemical staining, in matched HCC and adjacent liver samples from 178 patients with HCC after curative resection. The correlations of PAI1 H-scores with clinicopathologic variables and survival were further evaluated. Its prognostic value was finally confirmed in some public databases. RESULTS: It was found that PAI1 expression was significantly higher in HCC than in adjacent liver tissues. Moreover, high PAI1 expression was more frequent in those with multiple lesions. Univariate analyses showed that PAI1 expression was negatively associated with both overall and relapse-free survival. Although PAI1 expression was not statistically significant in multivariate Cox regression test, combination of it with TNM stage effectively distinguished survival and relapse, and served as an independent prognostic factor. In the online available datasets of HCC and liver cancer used, SERPINE1, the gene encoding PAI1, was also revealed to be prognostic. CONCLUSIONS: Our data suggested that high PAI1 expression was predictive for unfavorable biological behavior and long-term prognosis in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Carcinoma, Hepatocellular/pathology , Cohort Studies , Databases, Genetic , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Plasminogen Activator Inhibitor 1/genetics , Prognosis
6.
World J Urol ; 38(10): 2501-2511, 2020 Oct.
Article in English | MEDLINE | ID: mdl-31797075

ABSTRACT

PURPOSE: To investigate the prognostic role of expression of urokinase-type plasminogen activator system members, such as urokinase-type activator (uPA), uPA-receptor (uPAR), and plasminogen activator inhibitor-1 (PAI-1), in patients treated with radical prostatectomy (RP) for prostate cancer (PCa). METHODS: Immunohistochemical staining for uPA system was performed on a tissue microarray of specimens from 3121 patients who underwent RP. Cox regression analyses were performed to investigate the association of overexpression of these markers alone or in combination with biochemical recurrence (BCR). Decision curve analysis was used to assess the clinical impact of these markers. RESULTS: uPA, uPAR, and PAI-1 were overexpressed in 1012 (32.4%), 1271 (40.7%), and 1311 (42%) patients, respectively. uPA overexpression was associated with all clinicopathologic characteristics of biologically aggressive PCa. On multivariable analysis, uPA, uPAR, and PAI-1 overexpression were all three associated with BCR (HR: 1.75, p < 0.01, HR: 1.22, p = 0.01 and HR: 1.20, p = 0.03, respectively). Moreover, the probability of BCR increased incrementally with increasing cumulative number of overexpressed markers. Decision curve analysis showed that addition of uPA, uPAR, and PAI-1 resulted in a net benefit compared to a base model comparing standard clinicopathologic features across the entire threshold probability range. In subgroup analyses, overexpression of all three markers remained associated with BCR in patients with favorable pathologic characteristics. CONCLUSION: Overexpression of uPA, uPAR, and PAI-1 in PCa tissue were each associated with worse BCR. Additionally, overexpression of all three markers is informative even in patients with favorable pathologic characteristics potentially helping clinical decision-making regarding adjuvant therapy and/or intensified follow-up.


Subject(s)
Biomarkers, Tumor/physiology , Neoplasm Recurrence, Local/etiology , Plasminogen Activator Inhibitor 1/physiology , Prostatectomy , Prostatic Neoplasms/etiology , Prostatic Neoplasms/surgery , Receptors, Urokinase Plasminogen Activator/physiology , Urokinase-Type Plasminogen Activator/physiology , Aged , Biomarkers, Tumor/biosynthesis , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/epidemiology , Plasminogen Activator Inhibitor 1/biosynthesis , Prognosis , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/metabolism , Receptors, Urokinase Plasminogen Activator/biosynthesis , Retrospective Studies , Urokinase-Type Plasminogen Activator/biosynthesis
7.
Drug Discov Ther ; 13(5): 280-287, 2019.
Article in English | MEDLINE | ID: mdl-31723100

ABSTRACT

Few studies have investigated the molecular mechanisms of catheter failure (CF). Herein, we performed histological and molecular biological analyses of the catheter tip to demonstrate its potential as a resource for biological investigation. Additionally, we searched for risk factors for the development of inflammation and coagulation, which are pathological conditions clarified by biological analysis. The CF group included 30 failed catheters involving thrombus and subcutaneous edema identified by ultrasonography. The No-CF group included 26 catheters with no complications. The removed catheter tips were fixed for hematoxylin-eosin (HE) staining with the application of a real-time reverse transcriptase polymerase chain reaction for eukaryotic 18S ribosomal RNA (rRNA), interleukin 1ß, tumor necrosis factor α, tissue plasminogen activator, and plasminogen activator inhibitor 1 (SERPINE1). HE staining identified attached nuclear cells on the inner surfaces of both CF and No-CF catheters. The 18S rRNA was amplified in all samples. The expression level of SERPINE1 was significantly higher in the CF group than in the No-CF group (p = 0.01), whereas the expression levels of other genes did not differ between the groups. Symptoms of CF associated with the expression of SERPINE1 were analyzed. The catheter being in contact with blood vessels during placement was a suggested factor related to the high expression of SERPINE1 (p = 0.04). Catheter tips are a potential resource for biological investigation, and expression analysis of the attached cells can reflect the pathological condition of the catheterized tissue. Further studies using catheter tips are required to elucidate the molecular mechanisms of CF.


Subject(s)
Catheters/adverse effects , Equipment Failure Analysis/statistics & numerical data , Aged , Case-Control Studies , Female , Gene Expression , Humans , Leukocytes/metabolism , Male , Plasminogen Activator Inhibitor 1/biosynthesis
8.
Am J Pathol ; 189(6): 1284-1297, 2019 06.
Article in English | MEDLINE | ID: mdl-30953603

ABSTRACT

A vital constituent of the centrosome involved in regulating the activity of the organelle during the cell cycle is centrosomal protein (CEP)-72, whose function in the case of human cancer yet lacks clarity. The expression dynamics of CEP72 and its clinical impact were examined in a large cohort of bladder tissues. Several experiments at both the in vitro and in vivo levels on urothelial carcinoma of the bladder (UCB) cells were conducted to understand the role of this molecule along with the mechanisms. Overexpression of CEP72 in UCB was linked with the acquisition of an aggressive phenotype, which was associated with poor prognosis. In UCB cell lines, knockdown of CEP72 using shRNA was sufficient to inhibit cell invasiveness/metastasis, whereas ectopic overexpression of CEP72 promoted cell invasiveness and/or metastasis both in vitro and in vivo. CEP72 was demonstrated to induce UCB cell aggressiveness via up-regulation of an important target downstream, the serpin family member 1 gene (SERPINE1) (alias plasminogen activator inhibitor, PAI1), ultimately leading to increased cancer cell invasiveness. Particularly, overexpression of CEP72 was associated with a sizable increase in cAMP response element-binding protein binding at the SERPINE1 promoter, leading to increased SERPINE1 transcription. Such observations are suggestive of the potential use of CEP72 as a therapeutic tool for UCB.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder/metabolism , Aged , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
9.
Exp Mol Pathol ; 106: 1-6, 2019 02.
Article in English | MEDLINE | ID: mdl-30414981

ABSTRACT

Graves' ophthalmopathy (GO), a complication of Graves' disease (GD), is typified by orbital inflammation, ocular tissue expansion and remodeling and, ultimately, fibrosis. Orbital fibroblasts are key effectors of GO pathogenesis exhibiting exaggerated inflammatory and fibroproliferative responses to cytokines released by infiltrating immune cells. Activated orbital fibroblasts also produce inflammatory mediators that contribute to disease progression, facilitate the orbital trafficking of monocytes and macrophages, promote differentiation of matrix-producing myofibroblasts and stimulate accumulation of a hyaluronan-rich stroma, which leads to orbital tissue edema and fibrosis. Proteomic and transcriptome profiling of the genomic response of ocular and non-ocular fibroblasts to INF-γ and TGF-ß1 focused on identification of translationally-relevant therapeutic candidates. Induction of plasminogen activator inhibitor-1 (PAI-1, SERPINE1), a clade E member of the serine protease inhibitor (SERPIN) gene family and a prominent regulator of the pericellular proteolytic microenvironment, was one of the most highly up-regulated proteins in INF-γ- or TGF-ß1-stimulated GO fibroblasts as well as in severe active GD compared to patients without thyroid disease. PAI-1 has multifunctional roles in inflammatory and fibrotic processes that impact tissue remodeling, immune cell trafficking and survival as well as signaling through several receptor systems. This review focuses on the pathophysiology of the GO fibroblast and possible targets for effective drug therapy.


Subject(s)
Biomarkers/analysis , Fibroblasts/metabolism , Graves Ophthalmopathy/metabolism , Adipocytes/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Profiling , Genes, Immediate-Early , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/pathology , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Interferon-gamma/pharmacology , Orbit/pathology , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/physiology , Signal Transduction , Transforming Growth Factor beta1/pharmacology
10.
Medicine (Baltimore) ; 97(41): e12799, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30313110

ABSTRACT

One of the risk factors for vascular obstetric complications, such as intrauterine growth restriction (IUGR), is inherited thrombophilias. Nevertheless, routine screening for thrombophilias is not endorsed in pregnant women due to their low prevalence and conflicting results of published studies regarding the usefulness of screening in these patients. The cause of IUGR remains unknown in almost 1 quarter of cases. There are no published studies evaluating the association of inherited thrombophilias and IUGR in patients with IUGR of unknown origin. Understanding and preventing IUGR is an important public health concern, as IUGR has been associated with fetal mortality and neonatal morbidity, as well as adverse long-standing consequences. This study aimed to evaluate the prevalence of inherited thrombophilias in IUGR of unknown cause and to test the association between the inherited thrombophilias and IUGR of unknown cause.This study included 33 cases of IUGR of unknown cause tested for inherited thrombophilias and 66 controls individually matched for age, ethnicity, and smoking status.Patients with plasminogen activator inhibitor 1 (PAI-1) and methylenetetrahydrofolate reductase (MTHFR) had significantly higher odds for IUGR of unknown cause (P < .001 and P = .002, respectively) with OR 13.546 (CI 95% 3.79-48.37) and 8.139 (CI 95% 2.20-30.10), respectively. A positive association between other inherited thrombophilias (homozygous 20210 prothrombin gene mutation and homozygous factor V Leiden) and IUGR of unknown cause was also found, P = .096, OR 6.106 (CI 95% 0.72-51.30), although it was not statistically significant (P = .096, OR = 6.106, CI 95% 0.72-51.30).Our results indicate that PAI-1 and MTHFR thrombophilias represent risk factors for IUGR of otherwise unidentified cause.


Subject(s)
Fetal Growth Retardation/etiology , Methylenetetrahydrofolate Reductase (NADPH2)/biosynthesis , Plasminogen Activator Inhibitor 1/biosynthesis , Thrombophilia/complications , Thrombophilia/genetics , Adult , Body Mass Index , Case-Control Studies , Female , Humans , Pregnancy , Retrospective Studies
11.
Mol Biol Rep ; 45(6): 2835-2841, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30250995

ABSTRACT

COPD is a chronic airway inflammatory disease characterized mainly by neutrophil airway infiltrations. The neutrophil airway inflammation is mainly mediated through a key player like the pro-inflammatory cytokine IL-17A which is involved in the modulation of p53-fibrinolytic system. This study was undertaken to examine the molecular changes for the expressions of IL-17A and p53-fibrinolytic system in smokers with or without COPD. Blood and serum samples were collected from ten patients of smokers having COPD and ten samples from smokers without COPD and ten healthy control subjects. Western blot analyses were performed to evaluate the expressions of IL-17A, p53 and PAI-1. Apoptosis was assessed by immunoblot for cleaved caspase-3. In addition, FEV% was also determined of these patients. qRT-PCR was done to detect the gene expression study from the blood samples on p53-fibrinolytic components. A significant difference was found in the expression levels of IL-17A in smokers with COPD patient when compared to smokers without COPD and the control subjects. Similarly the smokers with COPD showed significant increase in the fibrinolytic component PAI-1 as well as in expression levels of p53 when compared to smokers without COPD and normal subjects. Increased cleaved caspase-3 may also promote apoptosis.The expression pattern of the IL-17A in chronic obstructive pulmonary distress syndrome samples was increased as compared of those of normal samples, and their main role in the regulation of and p53-fibrinolytic system makes these components as a predictive prominent component in smokers with COPD.


Subject(s)
Interleukin-17/biosynthesis , Pulmonary Disease, Chronic Obstructive/blood , Smoking/blood , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Apoptosis/physiology , Case-Control Studies , Female , Fibrinolysis , Humans , Interleukin-17/blood , Interleukin-17/genetics , Male , Middle Aged , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activator Inhibitor 1/genetics , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/genetics , Smoking/adverse effects , Smoking/genetics , Tumor Suppressor Protein p53/blood , Tumor Suppressor Protein p53/genetics , Urokinase-Type Plasminogen Activator/blood
12.
Am J Physiol Lung Cell Mol Physiol ; 315(2): L149-L161, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29644896

ABSTRACT

We have previously reported that miR-17~92 is critically involved in the pathogenesis of pulmonary hypertension (PH). We also identified two novel mR-17/20a direct targets, PDZ and LIM domain protein 5 (PDLIM5) and prolyl hydroxylase 2 (PHD2), and elucidated the signaling pathways by which PDLIM5 and PHD2 regulate functions of pulmonary artery smooth muscle cells (PASMCs). In addition, we have shown that plasminogen activator inhibitor-1 (PAI-1) is also downregulated in PASMCs that overexpress miR-17~92. However, it is unclear whether PAI-1 is a direct target of miR-17~92 and whether it plays a role in regulating the PASMC phenotype. In this study, we have identified PAI-1 as a novel target of miR-19a/b, two members of the miR-17~92 cluster. We found that the 3'-untranslated region (UTR) of PAI-1 contains a miR-19a/b binding site and that miR-19a/b can target this site to suppress PAI-1 protein expression. MiR-17/20a, two other members of miR-17~92, may also indirectly suppress PAI-1 expression through PDLIM5. PAI-1 is a negative regulator of miR-17~92-mediated PASMC proliferation. Silencing of PAI-1 induces Smad2/calponin signaling in PASMCs, suggesting that PAI-1 is a negative regulator of the PASMC contractile phenotype. We also found that PAI-1 is essential for the metabolic gene expression in PASMCs. Furthermore, although there is no significant change in PAI-1 levels in PASMCs isolated from idiopathic pulmonary arterial hypertension and associated pulmonary arterial hypertension patients, PAI-1 is downregulated in hypoxia/Sugen-induced hypertensive rat lungs. These results suggest that miR-17~92 regulates the PASMC contractile phenotype and proliferation coordinately and synergistically by direct and indirect targeting of PAI-1.


Subject(s)
MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Pulmonary Artery/metabolism , Signal Transduction , 3' Untranslated Regions , Animals , Cell Proliferation , Gene Expression Regulation , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Male , MicroRNAs/genetics , Muscle Contraction/genetics , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Plasminogen Activator Inhibitor 1/genetics , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley
13.
J Oral Pathol Med ; 47(6): 566-574, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29693741

ABSTRACT

BACKGROUND: In oral squamous cell carcinoma (OSCC), the HIF-1 complex promotes the expression of genes involved in specific mechanisms of cell survival under hypoxic conditions, such as plasminogen activator inhibitor-1 (PAI-1), carbonic anhydrase 9 (CAIX), and vascular endothelial growth factor A (VEGFA). The study aimed to investigate the presence and prognostic value of PAI-1, CAIX, and VEGFA in OSCC. MATERIALS AND METHODS: Immunohistochemistry was used to analyze the expressions of these proteins in 52 tumoral tissue samples of patients with OSCC, surgically treated and followed by a minimum of 24 months after surgery. The correlations between protein expressions and clinicopathological parameters and prognosis were analyzed. RESULTS: Positive PAI-1 membrane expression was significantly associated with local disease relapse (P = .027). Multivariate analysis revealed that the positive PAI-1 membrane expression is an independent marker for local disease relapse, with approximately 14-fold increased risk when compared to negative expression (OR = 14.49; CI = 1.40-150.01, P = .025). Strong PAI-1 cytoplasmic expression was significantly associated with the less differentiation grade (P = .027). Strong CAIX membrane expression was significantly associated with local disease-free survival (P = .038). Positive CAIX cytoplasmic expression was significantly associated with lymph node affected (P = .025) and with disease-specific survival (P = .022). Multivariate analysis revealed that the positive CAIX cytoplasmic expression is an independent risk factor for disease-related death, increasing their risk approximately 3-fold when compared to negative expression (HR = 2.84; CI = 1.02-7.87, P = .045). Positive VEGFA cytoplasmic expression was significantly associated with less differentiation grade (P = .035). CONCLUSION: Our results suggest a potential role for these expressions profiles as tumor prognostic markers in OSCC patients.


Subject(s)
Antigens, Neoplasm/biosynthesis , Biomarkers, Tumor/biosynthesis , Carbonic Anhydrase IX/biosynthesis , Mouth Neoplasms/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Squamous Cell Carcinoma of Head and Neck/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Aged , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Carbonic Anhydrase IX/metabolism , Disease-Free Survival , Female , Humans , Immunohistochemistry , Lymph Nodes/pathology , Male , Middle Aged , Mouth Neoplasms/pathology , Multivariate Analysis , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Plasminogen Activator Inhibitor 1/metabolism , Prognosis , Squamous Cell Carcinoma of Head and Neck/pathology , Survival Analysis , Vascular Endothelial Growth Factor A/metabolism
14.
Toxicol Appl Pharmacol ; 345: 48-56, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29524502

ABSTRACT

Benzo[a]pyrene (BaP) is an environmental pollutant found in cigarette smoke and is implicated as a causative agent of tobacco-related diseases, such as arteriosclerosis. In contrast, vitamin D signaling, which is principally mediated by conversion of vitamin D to the active form, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], decreases cardiovascular disease risk. However, combined treatment with BaP and 1,25(OH)2D3 enhances BaP toxicity, including BaP-DNA adduct formation. We further investigated the cross-talk between BaP and 1,25(OH)2D3 signaling pathways, and found that combined treatment with these compounds induces mRNA and protein expression of plasminogen activator inhibitor 1 (PAI-1) in monocyte/macrophage-derived THP-1 and U937 cells. Protein synthesis inhibitor treatment did not inhibit induction of the PAI-1 gene (SERPINE1) in these cells. BaP plus 1,25(OH)2D3 induced differentiation markers, inhibited cellular proliferation, and induced apoptosis and oxidative stress in these cells. Reactive oxygen species scavenger treatment suppressed apoptosis but not SERPINE1 induction in cells treated with BaP plus 1,25(OH)2D3. Thus, combined treatment with BaP and 1,25(OH)2D3 induced SERPINE1 mRNA expression in these cells through a mechanism that does not require de novo protein synthesis or reactive oxygen species production. These findings suggest that induction of the proinflammatory factor PAI-1 plays a role in BaP toxicity. Interestingly, PAI-1 knockdown decreased expression of the cell surface antigen CD14, a monocytic differentiation marker, in THP-1 cells treated with BaP plus 1,25(OH)2D3. PAI-1 induction may also be related to a function of monocytes/macrophages in response to xenobiotic and vitamin D signaling.


Subject(s)
Benzo(a)pyrene/administration & dosage , Cholecalciferol/administration & dosage , Macrophages/drug effects , Macrophages/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Cell Survival/drug effects , Cell Survival/physiology , Drug Combinations , Gene Expression , Hep G2 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Plasminogen Activator Inhibitor 1/genetics , THP-1 Cells/drug effects , THP-1 Cells/metabolism , U937 Cells
15.
Exp Biol Med (Maywood) ; 243(5): 437-443, 2018 03.
Article in English | MEDLINE | ID: mdl-29444596

ABSTRACT

Senescence of chondrocytes and cartilage degeneration induced by the proinflammatory cytokine interleukin-1ß is associated with the pathogenesis of osteoarthritis. The cannabinoid receptor 1 has been involved in the pathological development of various diseases. Here, we evaluated whether activation of cannabinoid receptor 1 using its selective agonist arachidonyl-2-chloroethylamide had an influence on cellular senescence induced by interleukin-1ßin human chondrocytes. Our findings demonstrate that agonist arachidonyl-2-chloroethylamidedecreased senescence-associated ß-galactosidase activity and cell cycle arrest in the G0/G1 phase induced by interleukin-1ß. Importantly, our results display interleukin-1ßtreatment significantly increased the expressions of senescence genes (caveolin-1, PAI-1 and p21), which were prevented by agonist arachidonyl-2-chloroethylamide treatment. However, it was noticed that these functions of agonist arachidonyl-2-chloroethylamide were abolished by the cannabinoid receptor 1 selective antagonist AM251, suggesting the involvement of cannabinoid receptor 1. Also, our results indicate that agonist arachidonyl-2-chloroethylamide enhanced the expression of sirt1. These findings suggest that activation of cannabinoid receptor 1 by agonist arachidonyl-2-chloroethylamide might have a protective effect against pro-inflammatory cytokines such as interleukin-1ß-induced chondrocytes senescence in osteoarthritis patients. Impact statement Senescence of chondrocytes and cartilage degeneration induced by the proinflammatory cytokine interleukin-1ß (IL-1ß) are associated with the pathogenesis of osteoarthritis (OA). Here we found that: (a) the CB1 agonist ACEA abolished IL-1ß-induced senescence and cell arrest in chondrocytes; (b) the CB1 agonist ACEA also abolished IL-1ß-induced expression of caveolin-1, PAI-1, and p21;


Subject(s)
Arachidonic Acids/pharmacology , Cellular Senescence/drug effects , Chondrocytes/metabolism , Osteoarthritis/pathology , Receptor, Cannabinoid, CB1/metabolism , Sirtuin 1/metabolism , Caveolin 1/biosynthesis , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Enzyme Activation/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Interleukin-1beta/metabolism , Male , Middle Aged , Piperidines/pharmacology , Plasminogen Activator Inhibitor 1/biosynthesis , Pyrazoles/pharmacology , beta-Galactosidase/metabolism
17.
J Cell Biochem ; 119(6): 4692-4710, 2018 06.
Article in English | MEDLINE | ID: mdl-29278662

ABSTRACT

This study aims to investigate the effects of microRNA-335-5p (miR-335-5p) on lower-extremity deep vein thrombosis (LEDVT) by targeting PAI-1 through the TLR4 signaling pathway in rat models. siRNA, mimic, and inhibitor were used for transfection. The miR-335-5p expression was detected by in situ hybridization. CCK-8 assay and flow cytometry were adopted to detect proliferation, cell cycle, and apoptosis, respectively. Scratch test and Matrigel-based tube formation assay were used to detect the effect of miR-335-5p on cell migration ability and tube formation ability. A miR-335-5p lentivirus plasmid was constructed and injected into LEDVT rats. The length and weight of thrombus were measured, changes of thrombus recanalization were observed by CD34 immunohistochemistry, and levels of PAI-1 and inflammatory factors in femoral vein blood were detected by ELISA. LEDVT rats showed a higher AOD value of PAI-1, higher expression of PAI-1, NF-κB, Rac1, IL-1ß, and TLR4 and a lower miR-335-5p expression. PAI-1 and miR-335-5p were negatively correlated. Compared to the blank and siRNA-NC groups, the miR-335-5p mimic and siRNA-PAI-1 groups showed declined expression of PAI-1, TLR4, NF-κB, Rac1, and IL-1ß, increased proliferation and tube formation abilities, less cells in G0/G1 phase, and decreased apoptosis, decreased length and weight of thrombus, organized thrombus, increased new blood vessels, and decreased levels of PAI-1, IL-1, IL-6, and Tnf-a. miR-335-5p may suppress the occurrence and development of LEDVT in rats by repressing the activation of the TLR4 signaling pathway by targeted inhibition of PAI-1.


Subject(s)
Hindlimb/blood supply , MicroRNAs/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Signal Transduction , Toll-Like Receptor 4/metabolism , Venous Thrombosis/metabolism , Animals , Female , G1 Phase , Gene Expression Regulation , Hindlimb/metabolism , Hindlimb/pathology , Male , Rats , Rats, Sprague-Dawley , Resting Phase, Cell Cycle , Venous Thrombosis/pathology
18.
J Pharmacol Exp Ther ; 364(3): 399-408, 2018 03.
Article in English | MEDLINE | ID: mdl-29263242

ABSTRACT

Intra-abdominal adhesion is a common complication after laparotomy. Conventional therapeutic strategies still cannot safely and effectively prevent this disorder. In this study, a combination of chitosan, cellulose, and seaweed polysaccharide (thereafter referred as CCS) was developed to significantly alleviate the formation of postoperative adhesion in rats with abdominal trauma. Transforming growth factor ß1 (TGF-ß1, an important promoter of fibrosis) and its downstream factors-namely, alpha-smooth muscle actin and plasminogen activator inhibitor-1 (PAI-1)-were effectively suppressed by CCS in vivo, and as a result, the activation of tissue plasminogen activator (tPA, may generate plasmin that is a fibrinolytic factor capable of breaking down fibrin) was significantly promoted, presenting antifibrosis effects of CCS. In addition, the activity of kinases [e.g., transforming growth factor-activated kinase 1 (TAK1), c-Jun N-terminal kinase (JNK)/Stress-activated Protein Kinase (SAPK), and p38] in the mitogen-activated protein kinase (MAPK) inflammation signaling pathway was also significantly suppressed by CCS in vivo, demonstrating anti-inflammatory functions of CCS. The histologic studies further confirmed the role of CCS in the inhibition of fibrosis, collagen deposition, inflammation, and vascular proliferation. These results indicate the clinical potential of CCS in the treatment of postoperative intra-abdominal adhesion. CCS may induce both antifibrosis and anti-inflammatory effects, potentially inhibiting the postoperative intra-abdominal adhesion. For antifibrosis effects, the expression of PAI-1 (a key factor for the adhesion formation) can be regulated by different TGF-ß1-associated signaling pathways, such as the Smads/p53 pathway, metalloproteinase/tissue inhibitor of matrix metalloproteinases pathway, Mitogen-activated Extracellular signal-regulated Kinase (MEK)/extracellular regulated protein kinase (ERK) pathway, and Yes-associated protein/transcriptional coactivator with PDZ-binding motif pathway. Following the downregulation of PAI-1 achieved by CCS, the activation of tPA (which may generate plasmin that is a fibrinolytic factor capable of breaking down fibrin) is significantly promoted. For anti-inflammation effects, CCS may suppress the phosphorylation of classic kinases (e.g., TAK1, JNK, and p38) in the MAPK signaling pathway. In addition to the MAPK pathway, inflammatory pathways, such as Nuclear Factor-κ-gene Binding(NF-κB), MEK/ERK, and Ras homologue protein/Rho associated coiled coil forming protein, are associated with the formation of intra-abdominal adhesion. Therefore, the prevention mechanisms of CCS will be further investigated in the future, with a hope of fully understanding of antiadhesion effects.


Subject(s)
Abdomen/pathology , Cellulose/pharmacology , Chitosan/pharmacology , Polysaccharides/pharmacology , Postoperative Complications/drug therapy , Seaweed/chemistry , Actins/biosynthesis , Animals , Cellulose/therapeutic use , Chitosan/therapeutic use , Collagen/metabolism , Drug Interactions , Male , Neovascularization, Pathologic/drug therapy , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/biosynthesis , Polysaccharides/therapeutic use , Postoperative Complications/metabolism , Rats , Rats, Wistar , Tissue Adhesions/drug therapy , Tissue Adhesions/metabolism , Transforming Growth Factor beta1/biosynthesis
19.
Drug Res (Stuttg) ; 68(3): 153-158, 2018 03.
Article in English | MEDLINE | ID: mdl-28992660

ABSTRACT

BACKGROUND: Alpha mangostin has been reported to have activity for the treatment of liver fibrosis in the rats. However, the mechanisms of action are poorly understood. This study was aimed to investigate the effect of alpha mangostin on hepatic stellate cells (HSC) activation and proliferation through TGF-ß/Smad and Akt signaling pathways. METHODS: Immortalized HSC, LX2 cells, were incubated with TGF-ß with or without alpha mangostin (5 or 10 µM). Sorafenib 10 µM was used as positive control. LX2 viability was counted using trypan blue exclusion method. The effect of alpha mangostin on TGF-ß concentrations, and the expressions of proliferation and fibrogenic markers were evaluated. RESULTS: Alpha mangostin treatment resulted in a reduced proliferation of HSC, decreased Ki-67 and p-Akt expressions. These findings were followed with decreased concentrations of TGF-ß in the medium of cells treated with alpha mangostin, decreased expressions of COL1A1, TIMP1, PAI1, α-SMA, and p-Smad3 as fibrogenic markers. These effects were shown to be dose-dependent. CONCLUSIONS: Alpha mangostin inhibits hepatic stellate cells proliferation and activation through TGF-ß/Smad and Akt signaling pathways in dose dependent manner.


Subject(s)
Actins/biosynthesis , Collagen Type I/biosynthesis , Hepatic Stellate Cells/drug effects , Plasminogen Activator Inhibitor 1/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Smad3 Protein/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Transforming Growth Factor beta/metabolism , Xanthones/pharmacology , Animals , Biomarkers/metabolism , Cell Proliferation , Cells, Cultured , Collagen Type I, alpha 1 Chain , Dose-Response Relationship, Drug , Ki-67 Antigen/biosynthesis , Male , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Rats , Signal Transduction/drug effects , Smad3 Protein/metabolism , Sorafenib
20.
J Cell Physiol ; 233(3): 2067-2074, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28294324

ABSTRACT

Fibrosis of the subsynovial connective tissue (SSCT) in carpal tunnel syndrome (CTS) patients is increasingly recognized as an important aspect of CTS pathophysiology. In this study, we evaluated the effect of blocking profibrotic pathways in fibroblasts from the SSCT in CTS patients. Fibroblasts were stimulated with transforming growth factor ß1 (TGF-ß1), and then treated either with a specific fibrosis pathway inhibitor targeting TGF-ß receptor type 1 (TßRI), platelet-derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR), or vascular endothelial growth factor receptor (VEGFR). Fibrosis array and quantitative real-time polymerase chain reaction of fibrotic genes were evaluated. Array gene expression analysis revealed significant down-regulation of multiple fibrotic genes after treatment with TßRI, PDGFR, and VEGFR inhibitors. No array fibrotic genes were significantly down-regulated with EGFR inhibition. Further gene expression analysis of known CTS fibrosis markers collagen type I A2 (Col1), collagen type III A1 (Col3), connective tissue growth factor (CTGF), and SERPINE1 showed significantly down-regulation after TßRI inhibition. In contrast, VEGFR inhibition significantly down-regulated CTGF and SERPINE1, whereas, PDGFR and EGFR inhibition significantly down-regulated Col3. Taken together the inhibition of TßRI appears to be the primary mediator of fibrotic gene expression in fibroblasts from CTS patients. TGF-ß/Smad activity was further evaluated, and as expected inhibition of Smad activity was significantly down-regulated after inhibition of TßRI, but not with PDGFR, VEGFR, or EGFR inhibition. These results indicate that local therapies specifically targeting TGF-ß signaling alone or in combination offer the potential of a novel local antifibrosis therapy for patients with CTS.


Subject(s)
Carpal Tunnel Syndrome/drug therapy , ErbB Receptors/antagonists & inhibitors , Fibrosis/pathology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Synovial Membrane/pathology , Transforming Growth Factor beta/metabolism , Carpal Tunnel Syndrome/pathology , Cells, Cultured , Collagen Type I/biosynthesis , Collagen Type I/genetics , Collagen Type III/biosynthesis , Collagen Type III/genetics , Connective Tissue/pathology , Connective Tissue Cells/cytology , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Fibroblasts/metabolism , Fibrosis/drug therapy , Humans , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Synovial Membrane/cytology
SELECTION OF CITATIONS
SEARCH DETAIL