Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.715
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Hum Reprod ; 27(6)2021 05 29.
Article in English | MEDLINE | ID: mdl-33881516

ABSTRACT

Plasma fibronectin 1 (FN1) levels are elevated in individuals with pre-eclampsia (PE), which may be applied as a possible b marker for vascular endothelial injury during PE. In the present study, the possible role of FN1 in the pathogenesis of PE and regulation of apoptosis and autophagy in vascular endothelial cells was explored. Plasma FN1 levels in 80 patients with PE and 40 healthy pregnant individuals were measured using ELISA to verify its relationship with the severity of PE. pcDNA3.1-FN1 or FN1-small interfering (si) RNA was used to manipulate the expression of FN1 in human umbilical vein endothelial cells (HUVECs) to assess the effects of FN1 on cell apoptosis, autophagy, and the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signaling pathway. It was found that upregulation of FN1 promoted apoptosis and autophagy, in addition to significantly inhibiting the activation of AKT and mTOR in HUVECs. By contrast, downregulation of FN1 expression inhibited cell apoptosis and autophagy, but increased AKT and mTOR phosphorylation in HUVECs that were cultured in serum samples obtained from patients with PE. Rescue experiments found that the PI3K/AKT inhibitor LY294002 reversed the effects of FN1-siRNA on apoptosis and autophagy in HUVECs cultured in serum from patients with PE. Therefore, data from the present study suggest that FN1 participates in the pathogenesis of PE by promoting apoptosis and autophagy in vascular endothelial cells, which is associated with the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Endothelial Cells/pathology , Fibronectins/physiology , Pre-Eclampsia/etiology , Adult , Apoptosis , Autophagy , Case-Control Studies , Chromones/pharmacology , Endothelial Cells/metabolism , Female , Fibronectins/biosynthesis , Fibronectins/blood , Fibronectins/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Genes, Reporter , Human Umbilical Vein Endothelial Cells , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Pre-Eclampsia/blood , Pre-Eclampsia/pathology , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Severity of Illness Index , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
2.
Anticancer Drugs ; 32(7): 693-702, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33675611

ABSTRACT

Lung squamous carcinoma (LUSC) is the second most frequent subtype of non-small cell lung cancer. Rarely gene alterations are identified in LUSC. Therefore, identifying LUSC-related genes to explain the relevant molecular mechanism is urgently needed. A potential biomarker, calcium-activated nucleotidase 1 (CANT1), was elevated in tissues of LUSC patients relative to normal cases based on the TCGA and/or GTEx database. CCK-8 and transwell tests were then implemented to measure the proliferative, invasive and migratory capacities, and showed that knockdown of CANT1 blocked LUSC cells proliferation. miR-607, predicted as an upstream factor for CANT1, was declined in LUSC using TargetScan analysis and luciferase activity test. Low miR-607 expression was related with unfavorable outcomes of LUSC patients. Moreover, miR-607 downregulation elevated cell viability, invasion and migration in LUSC cells, which was antagonized by si-CANT1. GEPIA website was accessed to estimate the relevance between CANT1 and epithelial-mesenchymal transition (EMT)-related positive factors. The protein levels of Fibronectin, Vimentin, Snail and ß-catenin were altered due to the abnormal CANT1 and miR-607 expression. Together, these data unveiled that miR-607/CANT1 pair may exert a vital role in the progression of LUSC through mediating EMT process, which would furnish an available therapeutic therapy for LUSC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , MicroRNAs/metabolism , Nucleotidases/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Survival , Down-Regulation , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Vimentin/biosynthesis
3.
Int J Mol Sci ; 22(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34298955

ABSTRACT

PURPOSE: The effects of Rho-associated coiled-coil containing protein kinase (ROCK) 1 and 2 inhibitor, ripasudil hydrochloride hydrate (Rip), ROCK2 inhibitor, KD025 or rosiglitazone (Rosi) on two-dimension (2D) and three-dimension (3D) cultured human conjunctival fibroblasts (HconF) treated by transforming growth factor (TGFß2) were studied. METHODS: Two-dimension and three-dimension cultured HconF were examined by transendothelial electrical resistance (TEER, 2D), size and stiffness (3D), and the expression of the extracellular matrix (ECM) including collagen1 (COL1), COL4 and COL6, fibronectin (FN), and α-smooth muscle actin (αSMA) by quantitative PCR (2D, 3D) in the presence of Rip, KD025 or Rosi. RESULTS: TGFß2 caused a significant increase in (1) the TEER values (2D) which were greatly reduced by Rosi, (2) the stiffness of the 3D organoids which were substantially reduced by Rip or KD025, and (3) TGFß2 induced a significant up-regulation of all ECMs, except for COL6 (2D) or αSMA (3D), and down-regulation of COL6 (2D). Rosi caused a significant up-regulation of COL1, 4 and 6 (3D), and down-regulation of COL6 (2D) and αSMA (3D). Most of these TGFß2-induced expressions in the 2D and αSMA in the 3D were substantially inhibited by KD025, but COL4 and αSMA in 2D were further enhanced by Rip. CONCLUSION: The findings reported herein indicate that TGFß2 induces an increase in fibrogenetic changes on the plane and in the spatial space, and are inhibited by Rosi and ROCK inhibitors, respectively.


Subject(s)
Conjunctiva/metabolism , Fibroblasts/metabolism , Rosiglitazone/pharmacology , Transforming Growth Factor beta2/metabolism , rho-Associated Kinases/antagonists & inhibitors , Actins/biosynthesis , Cell Line , Collagen/biosynthesis , Fibronectins/biosynthesis , Gene Expression Regulation/drug effects , Humans , rho-Associated Kinases/metabolism
4.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884812

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) recapitulates metastasis and can be induced in vitro through transforming growth factor (TGF)-ß signaling. A role for MMP activity in glioblastoma multiforme has been ascribed to EMT, but the molecular crosstalk between TGF-ß signaling and membrane type 1 MMP (MT1-MMP) remains poorly understood. Here, the expression of common EMT biomarkers, induced through TGF-ß and the MT1-MMP inducer concanavalin A (ConA), was explored using RNA-seq analysis and differential gene arrays in human U87 glioblastoma cells. TGF-ß triggered SNAIL and fibronectin expressions in 2D-adherent and 3D-spheroid U87 glioblastoma cell models. Those inductions were antagonized by the TGF-ß receptor kinase inhibitor galunisertib, the JAK/STAT inhibitors AG490 and tofacitinib, and by the diet-derived epigallocatechin gallate (EGCG). Transient gene silencing of MT1-MMP prevented the induction of SNAIL by ConA and abrogated TGF-ß-induced cell chemotaxis. Moreover, ConA induced STAT3 and Src phosphorylation, suggesting these pathways to be involved in the MT1-MMP-mediated signaling axis that led to SNAIL induction. Our findings highlight a new signaling axis linking MT1-MMP to TGF-ß-mediated EMT-like induction in glioblastoma cells, the process of which can be prevented by the diet-derived EGCG.


Subject(s)
Brain Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Glioblastoma/pathology , Matrix Metalloproteinase 14/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Snail Family Transcription Factors/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Concanavalin A , Fibronectins/biosynthesis , Humans , Matrix Metalloproteinase 14/genetics , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism , Tyrphostins/pharmacology
5.
Int J Mol Sci ; 22(7)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917452

ABSTRACT

Due to the increasing incidence of malignant gliomas, particularly glioblastoma multiforme (GBM), a simple and reliable GBM diagnosis is needed to screen early the death-threaten patients. This study aimed to identify a protein that can be used to discriminate GBM from low-grade astrocytoma and elucidate further that it has a functional role during malignant glioma progressions. To identify proteins that display low or no expression in low-grade astrocytoma but elevated levels in GBM, glycoprotein fibronectin (FN) was particularly examined according to the mining of the Human Protein Atlas. Web-based open megadata minings revealed that FN was mainly mutated in the cBio Cancer Genomic Portal but dominantly overexpressed in the ONCOMINE (a cancer microarray database and integrated data-mining platform) in distinct tumor types. Furthermore, numerous different cancer patients with high FN indeed exhibited a poor prognosis in the PrognoScan mining, indicating that FN involves in tumor malignancy. To investigate further the significance of FN expression in glioma progression, tumor specimens from five malignant gliomas with recurrences that received at least two surgeries were enrolled and examined. The immunohistochemical staining showed that FN expression indeed determined the distinct progressions of malignant gliomas. Furthermore, the expression of vimentin (VIM), a mesenchymal protein that is strongly expressed in malignant cancers, was similar to the FN pattern. Moreover, the level of epithelial-mesenchymal transition (EMT) inducer transforming growth factor-beta (TGF-ß) was almost recapitulated with the FN expression. Together, this study identifies a protein FN that can be used to diagnose GBM from low-grade astrocytoma; moreover, its expression functionally determines the malignant glioma progressions via TGF-ß-induced EMT pathway.


Subject(s)
Brain Neoplasms/metabolism , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Neoplasm Proteins/biosynthesis , Signal Transduction , Transforming Growth Factor beta/metabolism , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Databases, Nucleic Acid , Female , Fibronectins/genetics , Glioblastoma/diagnosis , Glioblastoma/diagnostic imaging , Glioblastoma/genetics , Humans , Male , Middle Aged , Neoplasm Proteins/genetics , Prognosis , Transforming Growth Factor beta/genetics
6.
J Biol Chem ; 294(20): 8218-8226, 2019 05 17.
Article in English | MEDLINE | ID: mdl-30952701

ABSTRACT

Excessive activation of the renin-angiotensin system (RAS) in diabetic cardiomyopathy (DCM) provokes a series of structural and functional abnormalities, and causes ventricular remodeling and heart failure in diabetes. (Pro)renin receptor (PRR) is a component of the RAS and has been reported to be up-regulated in some cardiovascular diseases. Furthermore, PRR blockade in some cardiovascular diseases, such as myocardial infarction and hypertension, has been demonstrated to reverse their pathogenesis. However, there have been few studies about the function of PRR in the pathogenesis of DCM. In this study, we hypothesized that PRR is involved in the pathogenesis of DCM and mediates myocardial injury in DCM. To explore the role of PRR in DCM, we evaluated the effects of PRR overexpression and knockdown on the DCM phenotype in vivo and in vitro The results show that PRR overexpression exacerbates myocardial injury and the inflammatory response in rats with DCM. Conversely, PRR knockdown alleviates myocardial fibrosis, apoptosis, and the inflammatory response, reversing the cardiac dysfunction in rats with DCM. In cell experiments, PRR overexpression also up-regulated the protein expression of collagen I and fibronectin, aggravated the inflammatory response, and increased the production of reactive oxygen species, whereas PRR knockdown had the opposite effect. Thus, PRR mediates myocardial injury, apoptosis, and the inflammatory response, likely through a PRR/extracellular signal-regulated kinase/reactive oxygen species pathway.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , MAP Kinase Signaling System , Myocardium/metabolism , Receptors, Cell Surface/metabolism , Renin-Angiotensin System , Animals , Collagen Type I/biosynthesis , Diabetes Mellitus, Experimental/pathology , Diabetic Cardiomyopathies/pathology , Fibronectins/biosynthesis , Fibrosis , Inflammation/metabolism , Inflammation/pathology , Male , Myocardium/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Prorenin Receptor
7.
Am J Physiol Cell Physiol ; 316(4): C509-C521, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30698990

ABSTRACT

The phosphorylation state of myosin regulatory light chain (MRLC) is central to the regulation of contractility that impacts cellular homeostasis and fate decisions. Rho-kinase (ROCK) and myosin light chain kinase (MLCK) are major kinases for MRLC documented to selectively regulate MRLC in a subcellular position-specific manner; specifically, MLCK in some nonmuscle cell types works in the cell periphery to promote migration, while ROCK does so at the central region to sustain contractility. However, it remains unclear whether or not the spatially selective regulation of the MRLC kinases is universally present in other cell types, including dedifferentiated vascular smooth muscle cells (SMCs). Here, we demonstrate the absence of the spatial regulation in dedifferentiated SMCs using both cell lines and primary cells. Thus, our work is distinct from previous reports on cells with migratory potential. We also observed that the spatial regulation is partly induced upon fibronectin stimulation and Krüppel-like factor 4 overexpression. To find clues to the mechanism, we reveal how the phosphorylation state of MRLC is determined within dedifferentiated A7r5 SMCs under the enzymatic competition among three major regulators ROCK, MLCK, and MRLC phosphatase (MLCP). We show that ROCK, but not MLCK, predominantly regulates the MRLC phosphorylation in a manner distinct from previous in vitro-based and in silico-based reports. In this ROCK-dominating cellular system, the contractility at physiological conditions was regulated at the level of MRLC diphosphorylation, because its monophosphorylation is already saturated. Thus, the present study provides insights into the molecular basis underlying the absence of spatial MRLC regulation in dedifferentiated SMCs.


Subject(s)
Cell Dedifferentiation/physiology , Fibronectins/biosynthesis , Kruppel-Like Transcription Factors/biosynthesis , Muscle, Smooth, Vascular/metabolism , Myosin Light Chains/physiology , Animals , Cattle , Cell Line , Cells, Cultured , Humans , Kruppel-Like Factor 4 , Myosin-Light-Chain Kinase/metabolism , Rats , rho-Associated Kinases/physiology
8.
Am J Physiol Renal Physiol ; 317(6): F1430-F1438, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31545926

ABSTRACT

Renal fibrosis is a key pathological phenomenon of chronic kidney disease (CKD) contributing to the progressive loss of renal function. UK383,367 is a procollagen C proteinase inhibitor that has been selected as a candidate for dermal antiscarring agents, whereas its role in renal fibrosis is unclear. In the present study, UK383,367 was applied to a CKD mouse model of unilateral ureteral obstruction (UUO) and cell lines of renal tubular epithelial cells (mouse proximal tubular cells) and renal fibroblast cells (NRK-49F cells) challenged by transforming growth factor-ß1. In vivo, bone morphogenetic protein 1, the target of UK383,367, was significantly enhanced in UUO mouse kidneys and renal biopsies from patients with CKD. Strikingly, UK383,367 administration ameliorated tubulointerstitial fibrosis as shown by Masson's trichrome staining in line with the blocked expression of collagen type I/III, fibronectin, and α-smooth muscle actin in the kidneys from UUO mice. Similarly, the enhanced inflammatory factors in obstructed kidneys were also blunted. In vitro, UK383,367 pretreatment inhibited the induction of collagen type I/III, fibronectin, and α-smooth muscle actin in both mouse proximal tubular cells and NRK-49F cells treated with transforming growth factor-ß1. Taken together, these findings indicate that the bone morphogenetic protein 1 inhibitor UK383,367 could serve as a potential drug in antagonizing CKD renal fibrosis by acting on the maturation and deposition of collagen and the subsequent profibrotic response and inflammation.


Subject(s)
Bone Morphogenetic Protein 1/antagonists & inhibitors , Oxadiazoles/therapeutic use , Renal Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Animals , Cell Line , Child , Child, Preschool , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Collagen Type III/antagonists & inhibitors , Collagen Type III/biosynthesis , Female , Fibronectins/antagonists & inhibitors , Fibronectins/biosynthesis , Fibrosis/drug therapy , Humans , Inflammation/pathology , Inflammation/prevention & control , Kidney/pathology , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/pathology , Ureteral Obstruction/complications
9.
Exp Eye Res ; 189: 107820, 2019 12.
Article in English | MEDLINE | ID: mdl-31589839

ABSTRACT

Elevated intraocular pressure (IOP) is a major risk factor for the development of primary open-angle glaucoma (POAG). This is from an increased aqueous humour (AH) outflow resistance through the trabecular meshwork (TM). The pathogenic mechanisms leading to the increase in TM outflow resistance are poorly understood but are thought to be from a dysregulation of the TM extracellular matrix (ECM) environment. ECM modification and turnover are crucial in regulating the resistance to aqueous outflow. ECM turnover is influenced by a complex interplay of growth factors such as transforming growth factors (TGFß) family and matrix metalloproteinases (MMPs). Elevated TGFß2 levels result in an increase in ECM deposition such as fibronectin leading to increased resistance. Fibronectin is a major component of TM ECM and plays a key role in its maintenance. Thrombospondins (TSP)-1 and -2 are important regulators of the ECM environment. TSP-1 has been implicated in the pathogenesis of POAG through activation of TGFß2 within the TM. TSP-2 does not contain the catalytic domain to activate latent TGFß, but is able to mediate the activities of MMP 2 and 9, thereby influencing ECM turnover. TSP-2 knock out mice show lower IOP levels compared to their wild type counterparts, suggesting the involvement of TSP-2 in the pathogenesis of POAG but its role in the pathogenesis of POAG remains unclear. The purpose of this study was to investigate the role of TSP-2 in trabecular meshwork ECM regulation and hence the pathogenesis of POAG. TSP-1 and TSP-2 expressions in immortalised glaucomatous TM cells (GTM3) and primary human non-glaucomatous (NTM) and glaucomatous cells (GTM) were determined by immunocytochemistry, immuno-blot analysis and qPCR following treatment with TGFß2 and Dexamethasone. The level of ECM protein fibronectin was determined in TM cells using immuno-blot analysis following treatment with TSP-1 or -2. TM cells secrete TSP-1 and -2 under basal conditions at the protein level and TSP-2 mRNA and protein levels were increased in response to TGFß2 three days post treatment. Exogenous treatment with TSP-2 up-regulated the expression of fibronectin protein in GTM3 cells, primary NTM and GTM cells. TSP-1 did not affect fibronectin protein levels in GTM3 cells. This suggests that the role of TSP-2 might be distinct from that of TSP-1 in the regulation of the TM cell ECM environment. TSP-2 may be involved in the pathogenesis of POAG and contribute to increased IOP levels by increasing the deposition of fibronectin within the ECM in response to TGFß2.


Subject(s)
Fibronectins/genetics , Gene Expression Regulation , Glaucoma, Open-Angle/genetics , Thrombospondins/genetics , Trabecular Meshwork/metabolism , Transforming Growth Factor beta2/metabolism , Up-Regulation , Cell Adhesion Molecules , Cells, Cultured , Fibronectins/biosynthesis , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Humans , Intraocular Pressure/physiology , RNA, Messenger/genetics , Thrombospondins/biosynthesis , Trabecular Meshwork/pathology
10.
Am J Respir Cell Mol Biol ; 58(3): 378-390, 2018 03.
Article in English | MEDLINE | ID: mdl-29019707

ABSTRACT

Glutaminolysis is the metabolic process of glutamine, aberration of which has been implicated in several pathogeneses. Although we and others recently found a diversity of metabolic dysregulation in organ fibrosis, it is unknown if glutaminolysis regulates the profibrotic activities of myofibroblasts, the primary effector in this pathology. In this study, we found that lung myofibroblasts demonstrated significantly augmented glutaminolysis that was mediated by elevated glutaminase 1 (Gls1). Inhibition of glutaminolysis by specific Gls1 inhibitors CB-839 and BPTES as well as Gls1 siRNA blunted the expression of collagens but not that of fibronectin, elastin, or myofibroblastic marker smooth muscle actin-α. We found that glutaminolysis enhanced collagen translation and stability, which were mediated by glutaminolysis-dependent mTOR complex 1 activation and collagen proline hydroxylation, respectively. Furthermore, we found that the amount of the glutaminolytic end product α-ketoglutarate (α-KG) was increased in myofibroblasts. Similar to glutaminolysis, α-KG activated mTOR complex 1 and promoted the expression of collagens but not of fibronectin, elastin, or smooth muscle actin-α. α-KG also remarkably inhibited collagen degradation in fibroblasts. Taken together, our studies identified a previously unrecognized mechanism by which a major metabolic program regulates the exuberant production of collagens in myofibroblasts and suggest that glutaminolysis is a novel therapeutic target for treating organ fibrosis, including idiopathic pulmonary fibrosis.


Subject(s)
Glutamine/metabolism , Ketoglutaric Acids/metabolism , Myofibroblasts/metabolism , Proline/chemistry , Pulmonary Fibrosis/pathology , TOR Serine-Threonine Kinases/metabolism , Actins/biosynthesis , Animals , Benzeneacetamides/pharmacology , Cells, Cultured , Collagen/biosynthesis , Disease Models, Animal , Elastin/biosynthesis , Enzyme Activation/physiology , Fibronectins/biosynthesis , Glutaminase/antagonists & inhibitors , Glutaminase/genetics , Glutaminase/metabolism , Humans , Hydroxylation , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/genetics , Sulfides/pharmacology , Thiadiazoles/pharmacology
11.
J Biol Chem ; 292(18): 7542-7553, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28320863

ABSTRACT

The aim of the study was to characterize the role of Rac1 GTPase for the mineralocorticoid receptor (MR)-mediated pro-fibrotic remodeling. Transgenic mice with cardiac overexpression of constitutively active Rac1 (RacET) develop an age-dependent phenotype with atrial dilatation, fibrosis, and atrial fibrillation. Expression of MR was similar in RacET and WT mice. The expression of 11ß hydroxysteroid dehydrogenase type 2 (11ß-HSD2) was age-dependently up-regulated in the atria and the left ventricles of RacET mice on mRNA and protein levels. Statin treatment inhibiting Rac1 geranylgeranylation reduced 11ß-HSD2 up-regulation. Samples of human left atrial myocardium showed a positive correlation between Rac1 activity and 11ß-HSD2 expression (r = 0.7169). Immunoprecipitation showed enhanced Rac1-bound 11ß-HSD2 relative to Rac1 expression in RacET mice that was diminished with statin treatment. Both basal and phorbol 12-myristate 13-acetate (PMA)-induced NADPH oxidase activity were increased in RacET and correlated positively with 11ß-HSD2 expression (r = 0.788 and r = 0.843, respectively). In cultured H9c2 cardiomyocytes, Rac1 activation with l-buthionine sulfoximine increased; Rac1 inhibition with NSC23766 decreased 11ß-HSD2 mRNA and protein expression. Connective tissue growth factor (CTGF) up-regulation induced by aldosterone was prevented with NSC23766. Cardiomyocyte transfection with 11ß-HSD2 siRNA abolished the aldosterone-induced CTGF up-regulation. Aldosterone-stimulated MR nuclear translocation was blocked by the 11ß-HSD2 inhibitor carbenoxolone. In cardiac fibroblasts, nuclear MR translocation induced by aldosterone was inhibited with NSC23766 and spironolactone. NSC23766 prevented the aldosterone-induced proliferation and migration of cardiac fibroblasts and the up-regulation of CTGF and fibronectin. In conclusion, Rac1 GTPase regulates 11ß-HSD2 expression, MR activation, and MR-mediated pro-fibrotic signaling.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2/biosynthesis , Endomyocardial Fibrosis/enzymology , Fibroblasts/enzymology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Neuropeptides/biosynthesis , Signal Transduction , rac1 GTP-Binding Protein/biosynthesis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Aldosterone/pharmacology , Animals , Cell Line , Connective Tissue Growth Factor/biosynthesis , Connective Tissue Growth Factor/genetics , Endomyocardial Fibrosis/pathology , Fibroblasts/pathology , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation/drug effects , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Mice, Mutant Strains , Myocardium/pathology , Myocytes, Cardiac/pathology , Neuropeptides/genetics , Rats , Rats, Sprague-Dawley , Sulfoxides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , rac1 GTP-Binding Protein/genetics
12.
J Cell Biochem ; 119(6): 4717-4728, 2018 06.
Article in English | MEDLINE | ID: mdl-29274284

ABSTRACT

Fibronectin 1 (FN1) is involved in cell adhesion and migration processes including embryogenesis, wound healing, blood coagulation, host defense, metastasis, and implicated in various biochemical processes. However, its effects on the development and progression of human cancer, especially colorectal cancer (CRC), are unclear. To evaluate the relationship between the expression of FN1 and the histopathologic parameters of patients with CRC or the proliferation, migration, and invasion of colorectal cancer cell lines, we screened FN1 as a new candidate gene which promotes development of CRC, in an independent dataset (The Human Protein Atlas website). Here, we reported that FN1 was elevated in CRC tissues compared with normal colon tissues. Further, FN1 expression level was correlated with age, lymph vascular invasion, and survival rate. Knockdown of FN1 in two CRC cell lines, LOVO, and SW1116, significantly inhibited cell proliferation, migration and invasion, and induced cell apoptosis. Western blot analysis showed that down-regulation of FN1 significantly decreased the expression of Bcl-2, MMP-9, Twist, and increased the expression of Bax, Caspase-3, and E-cadherin in LOVO and SW1116 cells. Then, we found that the protein ITGA5 was identified as a binding partner of FN1 and ITGA5 overexpression reversed FN1-induced tumorigenesis of CRC in vitro. Taken together, FN1 suppressed apoptosis and promoted viability, invasion, and migration in CRC through interacting with ITGA5. FN1 may be a prognostic factor and potential target for CRC treatment.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Down-Regulation , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Cell Transformation, Neoplastic/pathology , Colorectal Neoplasms/pathology , Female , HCT116 Cells , Humans , Male , Neoplasm Invasiveness , Tissue Array Analysis
13.
Toxicol Appl Pharmacol ; 356: 25-35, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30055191

ABSTRACT

Nickel as a heavy metal is known to bring threat to human health, and nickel exposure is associated with changes in fibroblast activation which may contribute to its fibrotic properties. H2S has recently emerged as an important gasotransmitter involved in numerous cellular signal transduction and pathophysiological responses. Interaction of nickel and H2S on fibroblast cell activation has not been studied so far. Here, we showed that a lower dose of nickel (200 µM) induced the activation of human fibroblast cells, as evidenced by increased cell growth, migration and higher expressions of α-smooth muscle actin (αSMA) and fibronectin, while high dose of nickel (1 mM) inhibited cell viability. Nickel reduced intracellular thiol contents and stimulated oxidative stress. Nickel also repressed the mRNA and protein expression of cystathionine gamma-lyase (CSE, a H2S-generating gene) and blocked the endogenous production of H2S. Exogenously applied NaHS (a H2S donor) had no effect on nickel-induced cell viability but significantly attenuated nickel-stimulated cell migration and the expression of αSMA and fibronectin. In contrast, CSE deficiency worsened nickel-induced αSMA expression. Moreover, H2S incubation reversed nickel-stimulated TGFß1/SMAD1 signal and blocked TGFß1-initiated expressions of αSMA and fibronectin. Nickel inhibited the interaction of Sp1 with CSE promoter but strengthened the binding of Sp1 with TGFß1 promoter, which was reversed by exogenously applied NaHS. These data reveal that H2S protects from nickel-stimulated fibroblast activation and CSE/H2S system can be a potential target for the treatment of tissue fibrosis induced by nickel.


Subject(s)
Fibroblasts/drug effects , Hydrogen Sulfide/pharmacology , Nickel/toxicity , Smad1 Protein/drug effects , Sp1 Transcription Factor/drug effects , Transforming Growth Factor beta1/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cystathionine gamma-Lyase/antagonists & inhibitors , Fibronectins/biosynthesis , Fibronectins/genetics , Humans , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , Zinc/metabolism
14.
Parasite Immunol ; 40(6): e12533, 2018 06.
Article in English | MEDLINE | ID: mdl-29719047

ABSTRACT

Cystic echinococcosis is characterized by fluid-filled hydatid cysts in the liver and lungs. The cysts are surrounded by a host fibrous layer (the pericyst) which acts to isolate the parasite from surrounding tissues. Previous studies in liver cysts have indicated that the parasite may be a stimulating fibrosis. The aim of this study was to investigate whether hydatid cyst fluid (HCF) could influence the potential for fibrosis to occur in lung tissue by stimulating epithelial to mesenchymal transition (EMT) in a human lung epithelial cell line. An adenocarcinoma-derived alveolar basal epithelial cell line (A549) was used as a model for human alveolar epithelial cells (AEC II). These were cultured in vitro with HCF (UK sheep origin). Assays to investigate cell proliferation, cell migration and expression of cytoskeletal markers showed that HCF could stimulate changes indicative of EMT, including enhanced cell proliferation and migration; increased expression of mesenchymal cytoskeletal markers (fibronectin and vimentin) accompanied by a down-regulation of an epithelial marker (E-cadherin). Molecules within hydatid cyst fluid are capable of inducing phenotypic changes in A549 cells indicating that the parasite has the potential to modify lung epithelial cells which could contribute to fibrotic reactions.


Subject(s)
Cyst Fluid/immunology , Echinococcosis/immunology , Echinococcus granulosus/immunology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/immunology , A549 Cells , Animals , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Cell Line , Cell Movement/physiology , Cell Proliferation/physiology , Cyst Fluid/parasitology , Cysts/parasitology , Echinococcosis/parasitology , Fibronectins/biosynthesis , Humans , Liver/parasitology , Liver/pathology , Liver Diseases/parasitology , Lung/cytology , Lung/parasitology , Lung/pathology , Respiratory Mucosa/cytology , Respiratory Mucosa/parasitology , Respiratory Mucosa/pathology , Sheep , Vimentin/biosynthesis
15.
Pharmacology ; 102(1-2): 1-8, 2018.
Article in English | MEDLINE | ID: mdl-29669350

ABSTRACT

BACKGROUND: Periplaneta americana is one of the ancient insect groups with the strongest vitality. Periplaneta americana extract (PAE) has been explored as an alternative remedy for many diseases. Although much progress has been made in the study about PAE, the role of the drug in renal disease is rarely reported, especially in renal fibrosis. This study was designed to evaluate the renoprotective effect of PAE treatment to renal fibrosis. METHOD: An in vivo, unilateral ureteral obstruction (UUO) mouse model was built. Then the mice were treated with PAE (100 mg/kg body weight) once daily by oral gavage, again starting on the day of UUO and continued for 1 week. At the end of 1 week, the mice were sacrificed; kidney samples were collected for further analysis. In vitro, Boston University mouse proximal tubular cells were plated in 35-mm dishes at a density of 0.3 * 106 cells/dish. Then the cells were treated with 5-ng/mL TGF-ß1 in serum-free DMEM medium for an indicated length of time. The experimental groups were pretreated with the indicated concentrations of PAE (0.3125 mg/mL). The cells were further cultured for 24 h, and then cells were monitored morphologically or collected for biochemical analyses. RESULTS: Both in vivo and vitro PAE inhibits the expression of FN and alpha-smooth muscle actin and suppresses renal fibrosis. Importantly, PAE protects against renal fibrosis by inhibiting Janus tyrosine kinase 2 (JAK)/signal transducer and activator of transcription 3 (STAT) tyrosine phosphorylation. CONCLUSION: PAE attenuates renal fibrosis through the suppression of the JAK2/STAT3 pathway.


Subject(s)
Fibrosis/prevention & control , Janus Kinase 2/antagonists & inhibitors , Kidney/pathology , Periplaneta , Plant Extracts/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Actins/biosynthesis , Animals , Cells, Cultured , Fibronectins/biosynthesis , Male , Mice , Phosphorylation/drug effects , Signal Transduction/drug effects
16.
Wiad Lek ; 71(3 pt 1): 474-478, 2018.
Article in English | MEDLINE | ID: mdl-29783208

ABSTRACT

OBJECTIVE: Introduction: The steady increase in the incidence of non-alcoholic steatohepatitis (NASH) on the background of obesity and chronic kidney disease (CKD) in people of working age in Ukraine. The aim: To establish the role of hydrogen sulfide in the mechanisms of mutual burden and progression of non-alcoholic steatohepatitis and chronic kidney disease in patients with obesity. PATIENTS AND METHODS: Materials and methods: 114 patients with NASH were examined on the background of obesity of І-ІІ degree, including: 52 patients with NASH (group 1) (without accompanying CKD), 62 patients with NASH with a comorbid CKD І-ІІ degree (group 2). The control group consisted of 20 practically healthy persons (PHPs) of the corresponding age and sex. RESULTS: Results: The obtained data testify that a significant increase in the synthesis of collagen and glycoproteins in patients with NASH, which arose on the background of obesity, that comorbid with CKD, which was accompanied by an ineffective resorption of newly formed collagen due to insufficient activation of collagenolysis and proteolysis, a significant imbalance in the system of connective tissue metabolism. CONCLUSION: Conclusions: A significant increase in the synthesis of collagen and glycoproteins (fibronectin) in patients with NASH, which was observed on the background of obesity, was established, which is accompanied by an ineffective resorption of newly formed collagen due to inhibition of collagenolysis against activation of proteinase inhibitors (α2-MG).


Subject(s)
Hydrogen Sulfide/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Renal Insufficiency, Chronic/etiology , Adult , Collagen/biosynthesis , Disease Progression , Female , Fibronectins/biosynthesis , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/complications , Obesity/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/metabolism , Ukraine
17.
Dev Biol ; 417(1): 11-24, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27435625

ABSTRACT

Primordial germ cells (PGCs) are a highly migratory cell population that gives rise to eggs and sperm. Much is known about PGC specification, but less about the processes that control PGC migration. In this study, we document a deficiency in PGC development in embryos carrying global homozygous null mutations in Msx1 and Msx2, both immediate downstream effectors of Bmp signaling pathway. We show that Msx1(-/-);Msx2(-/-) mutant embryos have defects in PGC migration as well as a reduced number of PGCs. These phenotypes are also evident in a Mesp1-Cre-mediated mesoderm-specific mutant line of Msx1 and Msx2. Since PGCs are not marked in Mesp1-lineage tracing, our results suggest that Msx1 and Msx2 function cell non-autonomously in directing PGC migration. Consistent with this hypothesis, we noted an upregulation of fibronectin, well known as a mediator of cell migration, in tissues through which PGCs migrate. We also noted a reduction in the expression of Wnt5a and an increase in the expression in Bmp4 in such tissues in Msx1(-/-);Msx2(-/-) mutants, both known effectors of PGC development.


Subject(s)
Cell Movement/genetics , Embryonic Germ Cells/cytology , Homeodomain Proteins/genetics , MSX1 Transcription Factor/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein 4/biosynthesis , Embryonic Germ Cells/metabolism , Fibronectins/biosynthesis , Homeodomain Proteins/metabolism , MSX1 Transcription Factor/metabolism , Mesoderm/cytology , Mice , Mice, Inbred BALB C , Mice, Knockout , Wnt-5a Protein/biosynthesis
18.
Cancer Sci ; 108(3): 419-426, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28064471

ABSTRACT

Vasohibin-2 (VASH2) is a homolog of VASH1, an endothelium-derived angiogenesis inhibitor. Vasohibin-2 is mainly expressed in cancer cells, and has been implicated in the progression of cancer by inducing angiogenesis and tumor growth. Although VASH2 has been recently reported to be involved in epithelial-mesenchymal transition (EMT), its precise roles are obscure. The aim of the present study was to clarify the role of VASH2 in the EMT of cancer cells in relation to transforming growth factor-ß (TGF-ß) signaling, which is a major stimulator of EMT. Decreased expression of VASH2 in ovarian cancer cells significantly repressed the expression of TGF-ß type I receptor, namely activin receptor-like kinase 5. Transforming growth factor-ß1-induced phosphorylation of Smad2 and Smad3 was markedly decreased in VASH2 knockdown cells while the expression of Smad2 and Smad3 was unchanged. Accordingly, the responses to TGF-ß1 shown by promoter assay and plasminogen activator inhibitor type 1 expression were significantly attenuated in VASH2 knockdown cells. Furthermore, knockdown of VASH2 in cancer cells abrogated the TGF-ß1-induced reduced expression of epithelial markers including E-cadherin, and the elevated expression of mesenchymal markers including fibronectin, ZEB2, and Snail2, suggesting that endogenous VASH2 is required for TGF-ß1-induced EMT. In accordance with these results, the effects of TGF-ß1 on cell morphology, migration, invasion, and MMP2 expression were also abrogated when VASH2 was knocked down. These results indicate that VASH2 played a significant role in the EMT by modulating the TGF-ß signaling. We propose that VASH2 would be a novel molecular target for the prevention of EMT in cancers.


Subject(s)
Angiogenic Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Ovarian Neoplasms/pathology , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Cadherins/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Female , Fibronectins/biosynthesis , Homeodomain Proteins/biosynthesis , Humans , Matrix Metalloproteinase 2/biosynthesis , Neovascularization, Pathologic/genetics , Ovarian Neoplasms/genetics , Phosphorylation/genetics , Plasminogen Activator Inhibitor 1/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/biosynthesis , Repressor Proteins/biosynthesis , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Snail Family Transcription Factors/biosynthesis , Zinc Finger E-box Binding Homeobox 2
19.
Int J Obes (Lond) ; 41(1): 137-148, 2017 01.
Article in English | MEDLINE | ID: mdl-27297797

ABSTRACT

BACKGROUND/OBJECTIVES: Myostatin (Mstn) has a pivotal role in glucose and lipid metabolism. Mstn deficiency leads to the increased browning of white adipose tissue (WAT), which results in the increased energy expenditure and protection against diet-induced obesity and insulin resistance. In this study, we investigated the molecular mechanism(s) through which Mstn regulates browning of white adipocytes. METHODS: Quantitative molecular analyses were performed to assess Mstn regulation of miR-34a and Fndc5 expression. miR-34a was overexpressed and repressed to investigate miR-34a regulation of Fndc5. Luciferase reporter analysis verified direct binding between miR-34a and the Fndc5 3'-untranslated region (UTR). The browning phenotype of Mstn-/- adipocytes was assessed through the analysis of brown fat marker gene expression, mitochondrial function and infrared thermography. The role of miR-34a and Fndc5 in this browning phenotype was verified through antibody-mediated neutralization of FNDC5, knockdown of Fndc5 by small interfering RNA and through miR-34a gain-of-function and loss-of-function experiments. RESULTS: Mstn treatment of myoblasts inhibited Fndc5 expression, whereas the loss of Mstn increased Fndc5 levels in muscles and in circulation. Mstn inhibition of Fndc5 is miR-34a dependent. Mstn treatment of C2C12 myoblasts upregulated miR-34a expression, whereas reduced miR-34a expression was noted in Mstn-/- muscle and WAT. Subsequent overexpression of miR-34a inhibited Fndc5 expression, whereas blockade of miR-34a increased Fndc5 expression in myoblasts. Reporter analysis revealed that miR-34a directly suppresses Fndc5 expression through a miR-34a-specific binding site within the Fndc5 3'UTR. Importantly, Mstn-mediated inhibition of Fndc5 was blocked upon miR-34a inhibition. Mstn-/- adipocytes showed reduced miR-34a, enhanced Fndc5 expression and increased thermogenic gene expression, which was reversed upon either neutralization of Fndc5 or Fndc5 knockdown. In agreement, Mstn-/- adipocytes have increased mitochondria, improved mitochondrial function and increased heat production. CONCLUSIONS: Mstn regulates Fndc5/Irisin expression and secretion through a novel miR-34a-dependent post-transcriptional mechanism. Loss of Mstn in mice leads to the increased Fndc5/Irisin expression, which contributes to the browning of white adipocytes.


Subject(s)
Adipocytes, White/metabolism , Adipose Tissue, Brown/metabolism , Fibronectins/metabolism , Gene Expression Regulation , MicroRNAs , Myostatin/metabolism , Signal Transduction , 3T3-L1 Cells , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fibronectins/biosynthesis , Fibronectins/genetics , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/metabolism , Thermogenesis
20.
Tumour Biol ; 39(3): 1010428317695921, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28351321

ABSTRACT

The phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway is crucial for tumor survival, proliferation, and progression, making it an attractive target for therapeutic intervention. In glioblastoma, activated mammalian target of rapamycin promotes invasive phenotype and correlates with poor patient survival. A wide range of mammalian target of rapamycin inhibitors are currently being evaluated for cytotoxicity and anti-proliferative activity in various tumor types but are not explored sufficiently for controlling tumor invasion and recurrence. We recently reported that mammalian target of rapamycin inhibitors-rapamycin, temsirolimus, torin 1, and PP242-suppressed invasion and migration promoted by tumor necrosis factor-alpha and phorbol-myristate-acetate in glioblastoma cells. As aggressive invasion and migration of tumors are associated with mesenchymal and stem-like cell properties, this study aimed to examine the effect of mammalian target of rapamycin inhibitors on these features in glioblastoma cells. We demonstrate that temsirolimus and torin 1 effectively reduced the constitutive as well as phorbol-myristate-acetate/oncostatin-M-induced expression of mesenchymal markers (fibronectin, vimentin, and YKL40) and neural stem cell markers (Sox2, Oct4, nestin, and mushashi1). The inhibitors significantly abrogated the neurosphere-forming capacity induced by phorbol-myristate-acetate and oncostatin-M. Furthermore, we demonstrate that the drugs dephosphorylated signal transducer and activator transcription factor 3, a major regulator of mesenchymal and neural stem cell markers implicating the role of signal transducer and activator transcription factor 3 in the inhibitory action of these drugs. The findings demonstrate the potential of mammalian target of rapamycin inhibitors as "stemness-inhibiting drugs" and a promising therapeutic approach to target glioma stem cells.


Subject(s)
Cell Proliferation/drug effects , Glioblastoma/drug therapy , Neoplasm Recurrence, Local/drug therapy , STAT3 Transcription Factor/genetics , TOR Serine-Threonine Kinases/genetics , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/drug effects , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Naphthyridines/administration & dosage , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neural Stem Cells/drug effects , Oncostatin M/administration & dosage , Phosphatidylinositol 3-Kinases , STAT3 Transcription Factor/biosynthesis , Signal Transduction/drug effects , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tetradecanoylphorbol Acetate/administration & dosage , Tetradecanoylphorbol Acetate/analogs & derivatives , Vimentin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL