Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Front Immunol ; 13: 1074099, 2022.
Article in English | MEDLINE | ID: mdl-36544756

ABSTRACT

CD95L (also known as FasL or CD178) is a member of the tumor necrosis family (TNF) superfamily. Although this transmembrane ligand has been mainly considered as a potent apoptotic inducer in CD95 (Fas)-expressing cells, more recent studies pointed out its role in the implementation of non-apoptotic signals. Accordingly, this ligand has been associated with the aggravation of inflammation in different auto-immune disorders and in the metastatic occurrence in different cancers. Although it remains to decipher all key factors involved in the ambivalent role of this ligand, accumulating clues suggest that while the membrane bound CD95L triggers apoptosis, its soluble counterpart generated by metalloprotease-driven cleavage is responsible for its non-apoptotic functions. Nonetheless, the metalloproteases (MMPs and ADAMs) involved in the CD95L shedding, the cleavage sites and the different stoichiometries and functions of the soluble CD95L remain to be elucidated. To better understand how soluble CD95L triggers signaling pathways from apoptosis to inflammation or cell migration, we propose herein to summarize the different metalloproteases that have been described to be able to shed CD95L, their cleavage sites and the biological functions associated with the released ligands. Based on these new findings, the development of CD95/CD95L-targeting therapeutics is also discussed.


Subject(s)
Neoplasms , fas Receptor , Humans , Fas Ligand Protein , Ligands , Metalloproteases/metabolism , Signal Transduction , Inflammation
2.
Front Artif Intell ; 4: 780843, 2021.
Article in English | MEDLINE | ID: mdl-35059637

ABSTRACT

In this paper we propose a new family of algorithms, ATENT, for training adversarially robust deep neural networks. We formulate a new loss function that is equipped with an additional entropic regularization. Our loss function considers the contribution of adversarial samples that are drawn from a specially designed distribution in the data space that assigns high probability to points with high loss and in the immediate neighborhood of training samples. Our proposed algorithms optimize this loss to seek adversarially robust valleys of the loss landscape. Our approach achieves competitive (or better) performance in terms of robust classification accuracy as compared to several state-of-the-art robust learning approaches on benchmark datasets such as MNIST and CIFAR-10.

3.
Cell Biol Int ; 44(5): 1142-1155, 2020 May.
Article in English | MEDLINE | ID: mdl-31965656

ABSTRACT

We sought to determine the mechanism by which angiotensin II (AngII) inhibits isoproterenol induced increase in adenylate cyclase (AC) activity and cyclic adenosine monophosphate (cAMP) production in bovine pulmonary artery smooth muscle cells (BPASMCs). Treatment with AngII stimulates protein kinase C-ζ (PKC-ζ), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and PKC-α activities, and also inhibits isoproterenol induced increase in AC activity and cAMP production in the cells. Pertussis toxin pretreatment eliminates AngII caused inhibition of isoproterenol induced increase in AC activity without a discernible change in PKC-ζ, NADPH oxidase, and PKC-α activities. Treatment of the cells with AngII increases α2 isoform of Gi (Giα2) phosphorylation; while pretreatment with chemical and genetic inhibitors of PKC-ζ and NADPH oxidase attenuate AngII induced increase in PKC-α activity and Giα2 phosphorylation, and also reverse AngII caused inhibition of isoproterenol induced increase in AC activity. Pretreatment of the cells with chemical and genetic inhibitors of PKC-α attenuate AngII induced increase in Giα2 phosphorylation and inhibits isoproterenol induced increase in AC activity without a discernible change in PKC-ζ and NADPH oxidase activities. Overall, PKCζ-NADPH oxidase-PKCα signaling axis plays a crucial role in Giα2 phosphorylation resulting in AngII-mediated inhibition of isoproterenol induced increase in AC activity in BPASMCs.


Subject(s)
Angiotensin II/pharmacology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidases/metabolism , Protein Kinase C/metabolism , Pulmonary Artery/cytology , Adenylyl Cyclases/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Cattle , Cell Culture Techniques , GTP-Binding Protein alpha Subunits/metabolism , Isoproterenol/pharmacology , Myocytes, Smooth Muscle/drug effects , Phosphorylation , Protein Kinase C-alpha/metabolism , Signal Transduction
4.
Dalton Trans ; 48(42): 16083-16090, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31616883

ABSTRACT

A recent experiment has revealed that additive free ester hydrogenation by Co-pincer complexes might follow an unusual non-bifunctional mechanism, however, the detailed mechanistic pathway is missing. It has been predicted that several intermediates and transition states are involved, having their essential role in the catalytic performances. Detailed theoretical studies are therefore essential in this regard for achieving more efficient ester hydrogenation catalysts. On the basis of first principles calculations, performed over Co(PNP)/(PNN) complexes, we present here the energetics and mechanistic details, showing the distinct orientations of different possible intermediates and transition states, and find the minimum energy pathway for the conversion of esters to alcohols. In the way, we find that some intermediates must undergo structural distortion for achieving the lowest potential energy barrier which must have a severe impact on the catalytic turnover frequency.

5.
J Cell Commun Signal ; 13(4): 473-489, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30661173

ABSTRACT

The signalling pathway involving MMP-2 and sphingosine-1-phosphate (S1P) in endothelin-1 (ET-1) induced pulmonary artery smooth muscle cell (PASMC) proliferation is not clearly known. We, therefore, investigated the role of NADPH oxidase derived O2.--mediated modulation of MMP2-sphingomyeline-ceramide-S1P signalling axis in ET-1 induced increase in proliferation of PASMCs. Additionally, protective role of the tea cathechin, epigallocatechin-3-gallate (EGCG), if any, in this scenario has also been explored. ET-1 markedly increased NADPH oxidase and MMP-2 activities and proliferation of bovine pulmonary artery smooth muscle cells (BPASMCs). ET-1 also caused significant increase in sphingomyelinase (SMase) activity, ERK1/2 and sphingosine kinase (SPHK) phosphorylations, and S1P level in the cells. EGCG inhibited ET-1 induced increase in SMase activity, ERK1/2 and SPHK phosphorylations, S1P level and the SMC proliferation. EGCG also attenuated ET-1 induced activation of MMP-2 by inhibiting NADPH oxidase activity upon inhibiting the association of the NADPH oxidase components, p47phox and p67phox in the cell membrane. Molecular docking study revealed a marked binding affinity of p47phox with the galloyl group of EGCG. Overall, our study suggest that ET-1 induced proliferation of the PASMCs occurs via NADPH oxidase-MMP2- Spm- Cer-S1P signalling axis, and EGCG attenuates ET-1 induced increase in proliferation of the cells by inhibiting NADPH oxidase activity.

6.
J Mol Model ; 24(9): 224, 2018 Aug 07.
Article in English | MEDLINE | ID: mdl-30088159

ABSTRACT

Hydride transfer is the most crucial step for the catalytic hydrogenation of CO2 in homogeneous condition. Here, we perform state-of-the-art calculations to show the effect of geometry and spin states of Ni-hydride complexes containing different types of multidentate phosphine ligands on their hydride transfer barrier. For doing this, we first choose Ni-bis(diphosphine) complexes of the type NiP4, which have been synthesized recently and then by extrapolating the idea we propose a new type of NiP2N2 complex showing much lower hydride transfer barrier. We also compute the hydricities of the Ni-hydride complexes in aqueous medium and try to correlate these thermodynamic quantities with the kinetic barrier of hydride transfer. Graphical Abstract NiP2N2 complex can efficiently hydrogenage CO2 with a quite low hydride transfer barrier.

7.
Nucleic Acids Res ; 45(22): 12681-12699, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29036586

ABSTRACT

Crosstalk between growth factors (GFs) and steroid hormones recurs in embryogenesis and is co-opted in pathology, but underlying mechanisms remain elusive. Our data from mammary cells imply that the crosstalk between the epidermal GF and glucocorticoids (GCs) involves transcription factors like p53 and NF-κB, along with reduced pausing and traveling of RNA polymerase II (RNAPII) at both promoters and bodies of GF-inducible genes. Essentially, GCs inhibit positive feedback loops activated by GFs and stimulate the reciprocal inhibitory loops. As expected, no alterations in DNA methylation accompany the transcriptional events instigated by either stimulus, but forced demethylation of regulatory regions broadened the repertoire of GF-inducible genes. We report that enhancers, like some promoters, are poised for activation by GFs and GCs. In addition, within the cooperative interface of the crosstalk, GFs enhance binding of the GC receptor to DNA and, in synergy with GCs, promote productive RNAPII elongation. Reciprocally, within the antagonistic interface GFs hyper-acetylate chromatin at unmethylated promoters and enhancers of genes involved in motility, but GCs hypoacetylate the corresponding regions. In conclusion, unmethylated genomic regions that encode feedback regulatory modules and differentially recruit RNAPII and acetylases/deacetylases underlie the crosstalk between GFs and a steroid hormone.


Subject(s)
Epidermal Growth Factor/pharmacology , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Promoter Regions, Genetic/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , DNA Methylation , Dexamethasone/pharmacology , Humans , NF-kappa B/metabolism , Protein Processing, Post-Translational/drug effects , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Arch Biochem Biophys ; 633: 1-14, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28822840

ABSTRACT

Treatment of human pulmonary artery smooth muscle cells (HPASMCs) with the thromboxane A2 receptor antagonist, SQ29548 inhibited U46619 stimulation of phospholipase D (PLD) and NADPH oxidase activities in the cell membrane. Pretreatment with apocynin inhibited U46619 induced increase in NADPH oxidase activity. The cell membrane contains predominantly PLD2 along with PLD1 isoforms of PLD. Pretreatment with pharmacological and genetic inhibitors of PLD2, but not PLD1, attenuated U46619 stimulation of NADPH oxidase activity. U46619 stimulation of PLD and NADPH oxidase activities were insensitive to BFA and Clostridium botulinum C3 toxin; however, pretreatment with secinH3 inhibited U46619 induced increase in PLD and NADPH oxidase activities suggesting a major role of cytohesin in U46619-induced increase in PLD and NADPH oxidase activities. Arf-1, Arf-6, cytohesin-1 and cytohesin-2 were observed in the cytosolic fraction, but only Arf-6 and cytohesin-1 were translocated to the cell membrane upon treatment with U46619. Coimmunoprecipitation study showed association of Arf-6 with cytohesin-1 in the cell membrane fraction. In vitro binding of GTPγS with Arf-6 required the presence of cytohesin-1 and that occurs in BFA insensitive manner. Overall, BFA insensitive Arf6-cytohesin1 signaling axis plays a pivotal role in U46619-mediated activation of PLD leading to stimulation of NADPH oxidase activity in HPASMCs.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP-Ribosylation Factors/genetics , Guanine Nucleotide Exchange Factors/genetics , NADPH Oxidases/genetics , Phospholipase D/genetics , Vasoconstrictor Agents/pharmacology , ADP Ribose Transferases/pharmacology , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/metabolism , Acetophenones/pharmacology , Antioxidants/pharmacology , Botulinum Toxins/pharmacology , Brefeldin A/pharmacology , Bridged Bicyclo Compounds, Heterocyclic , Cell Membrane/drug effects , Cell Membrane/metabolism , Fatty Acids, Unsaturated , GTPase-Activating Proteins/antagonists & inhibitors , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Hydrazines/pharmacology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism , Phospholipase D/antagonists & inhibitors , Phospholipase D/metabolism , Primary Cell Culture , Protein Synthesis Inhibitors/pharmacology , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/antagonists & inhibitors , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Signal Transduction , Triazoles/pharmacology
9.
Mol Cell Biochem ; 427(1-2): 111-122, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28013477

ABSTRACT

Matrix metalloproteinases (MMPs) play a crucial role in developing different types of lung diseases, e.g., pulmonary arterial hypertension (PAH). Green tea polyphenolic catechins such as EGCG and ECG have been shown to ameliorate various types of diseases including PAH. Our present study revealed that among the four green tea catechins (EGCG, ECG, EC, and EGC), EGCG and ECG inhibit pro-/active MMP-2 activities in pulmonary artery smooth muscle cell (PASMC) culture supernatant. Based on the above, we investigated the interactions of pro-/active MMP-2 with the green tea catechins by computational methods. In silico analysis revealed a strong interaction of pro-/active MMP-2 with EGCG/ECG, and galloyl group has been observed to be responsible for this interaction. The in silico analysis corroborated our experimental observation that EGCG and ECG are active in preventing both the proMMP-2 and MMP-2 activities. Importantly, these two catechins appeared to be better inhibitors for proMMP-2 in comparison to MMP-2 as revealed by gelatin zymogram and also by molecular docking studies. In many type of cells, activation of proMMP-2 occurs via an increase in the level of MT1-MMP (MMP-14). We, therefore, determined the interactions of MT1-MMP with the green tea catechins by molecular docking analysis. The study revealed a strong interaction of MT1-MMP with EGCG/ECG, and galloyl group has been observed to be responsible for the interaction.


Subject(s)
Catechin , Enzyme Precursors , Gelatinases , Matrix Metalloproteinase 2 , Molecular Docking Simulation , Protease Inhibitors , Tea/chemistry , Animals , Catechin/chemistry , Catechin/pharmacology , Cattle , Enzyme Precursors/antagonists & inhibitors , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Gelatinases/antagonists & inhibitors , Gelatinases/chemistry , Gelatinases/metabolism , Humans , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology
10.
Biomed Pharmacother ; 84: 340-347, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27668533

ABSTRACT

Green tea polyphenolic catechins have been shown to prevent various types of diseases such as pulmonary hypertension (PAH), cancer and cardiac and neurological disorders. Matrix metalloproteinases (MMPs) play an important role in the development of PAH. The present study demonstrated that among the four green tea catechins (EGCG, ECG, EC and EGC), EGCG and ECG inhibit pro-/active MMP-9 activities in pulmonary artery smooth muscle cell culture supernatant. Based on the above, we investigated the interactions of pro-/active MMP-9 with the green tea catechins by computational methods. In silico molecular docking analysis revealed a strong interaction between pro-/active MMP-9 and EGCG/ECG, and galloyl group appears to be responsible for this enhanced interaction. The molecular docking studies corroborate our experimental observation that EGCG and ECG are mainly active in preventing both the proMMP-9 and MMP-9 activities.


Subject(s)
Catechin/pharmacology , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Molecular Docking Simulation , Tea/chemistry , Animals , Binding Sites , Catechin/chemistry , Cattle , Cells, Cultured , Humans , Ligands , Matrix Metalloproteinase Inhibitors/chemistry , Polyphenols/chemistry , Polyphenols/pharmacology
11.
Arch Biochem Biophys ; 603: 91-101, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27210740

ABSTRACT

The aim of the present study is to establish the mechanism associated with the proliferation of PASMCs under ANG II stimulation. The results showed that treatment of PASMCs with ANG II induces an increase in cell proliferation and 100 nM was the optimum concentration for maximum increase in proliferation of the cells. Pretreatment of the cells with AT1, but not AT2, receptor antagonist inhibited ANG II induced cell proliferation. Pretreatment with pharmacological and genetic inhibitors of sphingomyelinase (SMase) and sphingosine kinase (SPHK) prevented ANG II-induced cell proliferation. ANG II has also been shown to induce SMase activity, SPHK phosphorylation and S1P production. In addition, ANG II caused an increase in proMMP-2 expression and activation, ERK1/2 phosphorylation and NADPH oxidase activation. Upon inhibition of MMP-2, SMase activity and S1P level were curbed leading to inhibition of cell proliferation. SPHK was phosphorylated by ERK1/2 during ET-1 stimulation of the cells. ANG II-induced ERK1/2 phosphorylation and proMMP-2 expression and activation in the cells were abrogated upon inhibition of NADPH oxidase activity. Overall, NADPH oxidase plays an important role in proMMP-2 expression and activation and that MMP-2 mediated SMC proliferation occurs through the involvement of Spm-Cer-S1P signaling axis under ANG II stimulation of PASMCs.


Subject(s)
Angiotensin II/pharmacology , Ceramides/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , Myocytes, Smooth Muscle/metabolism , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Sphingomyelins/metabolism , Animals , Cattle , Cell Proliferation , Lung/metabolism , NADPH Oxidases/metabolism , Oxygen/metabolism , Phosphorylation , Pulmonary Artery/cytology , RNA, Small Interfering/metabolism , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism , Transfection
12.
Mol Cell Biochem ; 415(1-2): 13-28, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26910780

ABSTRACT

Treatment of bovine pulmonary artery smooth muscle cells with endothelin-1 (ET-1) caused an increase in the expression and activation of proMMP-2 in the cells. The present study was undertaken to determine the underlying mechanisms involved in this scenario. We demonstrated that (i) pretreatment with NADPH oxidase inhibitor, apocynin; PKC-α inhibitor, Go6976; p(38)MAPK inhibitor SB203580 and NF-κB inhibitor, Bay11-7082 inhibited the expression and activation of proMMP-2 induced by ET-1; (ii) ET-1 treatment to the cells stimulated NADPH oxidase and PKCα activity, p(38)MAPK phosphorylation as well as NF-κB activation by translocation of NF-κBp65 subunit from cytosol to the nucleus, and subsequently by increasing its DNA-binding activity; (iii) ET-1 increases MT1-MMP expression, which was inhibited upon pretreatment with apocynin, Go6976, SB293580, and Bay 11-7082; (iv) ET-1 treatment to the cells downregulated TIMP-2 level. Although apocynin and Go6976 pretreatment reversed ET-1 effect on TIMP-2 level, yet pretreatment of the cells with SB203580 and Bay 11-7082 did not show any discernible change in TIMP-2 level by ET-1. Overall, our results suggest that ET-1-induced activation of proMMP-2 is mediated via cross-talk between NADPH oxidase-PKCα-p(38)MAPK and NFκB-MT1MMP signaling pathways along with a marked decrease in TIMP-2 expression in the cells.


Subject(s)
Endothelin-1/metabolism , Enzyme Precursors/metabolism , Gelatinases/metabolism , Matrix Metalloproteinase 14/metabolism , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Pulmonary Artery/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cattle , Cells, Cultured , Down-Regulation , Enzyme Activation , Enzyme Precursors/genetics , Enzyme Precursors/isolation & purification , Gelatinases/genetics , Gelatinases/isolation & purification , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/enzymology , Tissue Inhibitor of Metalloproteinase-2/metabolism
13.
Biomed Pharmacother ; 78: 50-59, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26898424

ABSTRACT

Tea is the most popular beverages all over the world. Polyphenols are found ubiquitously in tea leaves and their regular consumption has been associated with a reduced risk of a number of chronic diseases including cancer, cardiovascular and neurodegenerative diseases. Epigallocatechin-3-gallate (EGCG) is the most abundant polyphenol in tea leaves and received great attention due to their protective role in the prevention of the diseases. Rather than eliciting direct antioxidant effects, the mechanisms by which tea polyphenol express these beneficial properties appear to involve their interaction with cellular signaling pathways and related machinery that mediate cell function under both normal and pathological conditions. The central focus of this review is to provide an overview of the role that the major tea polyphenol, EGCG plays in preventing cancer, cardiovascular and neurodegenerative diseases. This review present epidemiological data, human intervention study findings, as well as animal and in vitro studies in support of these actions and delineates the molecular mechanism associated with the action of EGCG in ameliorating of such diseases.


Subject(s)
Catechin/analogs & derivatives , Disease , Health , Protective Agents/pharmacology , Animals , Catechin/pharmacology , Humans , Models, Biological
14.
Cell Biochem Funct ; 33(7): 463-77, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26486270

ABSTRACT

During remodelling of pulmonary artery, marked proliferation of pulmonary artery smooth muscle cells (PASMCs) occurs, which contributes to pulmonary hypertension. Thromboxane A2 (TxA2) has been shown to produce pulmonary hypertension. The present study investigates the inhibitory effect of epigallocatechin-3-gallate (EGCG) on the TxA2 mimetic, U46619-induced proliferation of PASMCs. U46619 at a concentration of 10 nM induces maximum proliferation of bovine PASMCs. Both pharmacological and genetic inhibitors of p(38)MAPK, NF-κB and MMP-2 significantly inhibit U46619-induced cell proliferation. EGCG markedly abrogate U46619-induced p(38)MAPK phosphorylation, NF-κB activation, proMMP-2 expression and activation, and also the cell proliferation. U46619 causes an increase in the activation of sphingomyelinase (SMase) and sphingosine kinase (SPHK) and also increase sphingosine 1 phosphate (S1P) level. U46619 also induces phosphorylation of ERK1/2, which phosphorylates SPHK leading to an increase in S1P level. Both pharmacological and genetic inhibitors of SMase and SPHK markedly inhibit U46619-induced cell proliferation. Additionally, pharmacological and genetic inhibitors of MMP-2 markedly abrogate U46619-induced SMase activity and S1P level. EGCG markedly inhibit U46619-induced SMase activity, ERK1/2 and SPHK phosphorylation and S1P level in the cells. Overall, Sphingomyeline-Ceramide-Sphingosine-1-phosphate (Spm-Cer-S1P) signalling axis plays an important role in MMP-2 mediated U46619-induced proliferation of PASMCs. Importantly, EGCG inhibits U46619 induced increase in MMP-2 activation by modulating p(38)MAPK-NFκB pathway and subsequently prevents the cell proliferation.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Cardiovascular Agents/pharmacology , Catechin/analogs & derivatives , Cell Proliferation/drug effects , Pulmonary Artery/cytology , Vasoconstrictor Agents/pharmacology , Animals , Cardiovascular Diseases/prevention & control , Catechin/pharmacology , Cattle , Cell Culture Techniques , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase Inhibitors , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects
15.
Mol Cell Biochem ; 400(1-2): 107-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25399298

ABSTRACT

Endothelin-1 (ET-1) is known as the most potent vasoconstrictor yet described. Infusion of ET-1 into isolated rabbit lung has been shown to cause pulmonary vasoconstriction with the involvement of arachidonic acid metabolites. Given the potency of arachidonic acid metabolites, the activity of phospholipase A2 must be tightly regulated. Herein, we determined the mechanisms by which ET-1 stimulates cPLA2 activity during ET-1 stimulation of bovine pulmonary artery smooth muscle cells. We demonstrated that (i) treatment of bovine pulmonary artery smooth muscle cells with ET-1 stimulates cPLA2 activity in the cell membrane; (ii) ET-1 caused increase in O 2 (·-) production occurs via NADPH oxidase-dependent mechanism; (iii) ET-1-stimulated NADPH oxidase activity is markedly prevented upon pretreatment with PKC-ζ inhibitor, indicating that PKC-ζ plays a prominent role in this scenario; (iv) ET-1-induced NADPH oxidase-derived O 2 (·-) stimulates an aprotinin sensitive protease activity due to prominent increase in [Ca(2+)]i; (v) the aprotinin sensitive protease plays a pivotal role in activating PKC-α, which in turn phosphorylates p(38)MAPK and subsequently Giα leading to the activation of cPLA2. Taken together, we suggest that cross-talk between p(38)MAPK and Giα with the involvement of PKC-ζ, NADPH oxidase-derived O 2 (·-) , [Ca(2+)]i, aprotinin-sensitive protease and PKC-α play a pivotal role for full activation of cPLA2 during ET-1 stimulation of pulmonary artery smooth muscle cells.


Subject(s)
Endothelin-1/metabolism , GTP-Binding Proteins/metabolism , Phospholipases A2/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Arachidonic Acid/metabolism , Cattle , Cell Membrane , Endothelin-1/genetics , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism , Phospholipases A2/biosynthesis , Pulmonary Artery/metabolism , Vasoconstriction/genetics , p38 Mitogen-Activated Protein Kinases/biosynthesis
16.
Mol Cell Biochem ; 385(1-2): 53-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24065388

ABSTRACT

We investigated the mechanism by which TxA2 mimetic, U46619, activates proMMP-2 in bovine pulmonary artery smooth muscle cells. Our study showed that treatment of the cells with U46619 caused an increase in the expression and subsequently activation of proMMP-2 in the cells. Pretreatment with p(38)MAPK inhibitor, SB203580; and NF-κB inhibitor, Bay11-7082 inhibited the expression and activation of proMMP-2 induced by U46619. U46619 also induced increase in MT1-MMP expression, which was inhibited upon pretreatment with SB203580 and Bay11-7082. U46619 treatment to the cells stimulated p(38)MAPK activity as well as NF-κB activation by IκB-α phosphorylation, translocation of NF-κBp65 subunit from cytosol to nucleus and subsequently, by increasing its DNA-binding activity. Induction of NF-κB activation seems to be mediated through IKK, as transfection of cells with either IKKα or IKKß siRNA prevented U46619-induced phosphorylation of IκB-α and NF-κBp65 DNA-binding activity. U46619 treatment to the cells also downregulated the TIMP-2 level. Pretreatment of the cells with SB203580 and Bay11-7082 did not show any discernible change in TIMP-2 level by U46619. Overall, U46619-induced activation of proMMP-2 is mediated via involvement of p(38)MAPK-NFκB-MT1MMP signaling pathway with concomitant downregulation of TIMP-2 expression in bovine pulmonary artery smooth muscle cells.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Enzyme Precursors/metabolism , Gelatinases/metabolism , Matrix Metalloproteinase 14/metabolism , Myocytes, Smooth Muscle/enzymology , NF-kappa B/metabolism , Pulmonary Artery/cytology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cattle , Down-Regulation/drug effects , Enzyme Activation/drug effects , I-kappa B Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-2/metabolism , Up-Regulation/drug effects
17.
Indian J Biochem Biophys ; 51(6): 449-56, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25823216

ABSTRACT

Aneurysms develop as a result of chronic inflammation of vascular bed, where progressive destruction of structural proteins, especially elastin and collagen of smooth muscle cells has been shown to manifest. The underlying mechanisms are an increase in local production of proinflammatory cytokines and subsequent increase in proteases, especially matrix metalloproteinases (MMPs) that degrade the structural proteins. The plasminogen system: urokinase-type PA (u-PA), tissue-type PA (t-PA) and plasminogen activator inhibitor-1 (PAI-1) and the MMPs system-MMPs and TIMPs contribute to the progression and development of aneurysms. Recent studies suggest that aneurysms may be genetically determined. To date, most observable candidate genes for aneurysm (elastin, collagen, fibrillin, MMPs and TIMPs) have been explored with little substantiation of the underlying cause and effect. Recently, overexpression of the MMP-2 gene has been suggested as an important phenomenon for aneurysm formation. Along with MMPs, matrix formation also depends on JNK (c-Jun N-terminal kinase) as its activation plays important role in downregulating several genes of matrix production. Under stress, activation of JNK by various stimuli, such as angiotensin II, tumor necrosis factor-α and interleukin-1ß has been noted significantly in vascular smooth muscle cells. Several therapeutic indications corroborate that inhibition of MMP-2 and JNK is useful in preventing progression of vascular aneurysms. This review deals with the role of proteases in the progression of vascular aneurysm.


Subject(s)
Aneurysm/immunology , Blood Vessels/immunology , Cytokines/immunology , Models, Cardiovascular , Models, Immunological , Peptide Hydrolases/immunology , Signal Transduction/immunology , Animals , Enzyme Activation , Humans
18.
Arch Biochem Biophys ; 540(1-2): 133-44, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24184446

ABSTRACT

Treatment of bovine pulmonary artery smooth muscle cells (BPASMCs) with U46619 attenuated isoproterenol caused stimulation of adenyl cyclase activity and cAMP production. Pretreatment with SQ29548 (Tp receptor antagonist), apocynin (NADPH oxidase inhibitor) and Go6976 (PKC-α inhibitor) eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity. Pretreatment with SQ29548 and apocynin prevented U46619 induced increase in NADPH oxidase activity, PKC-α activity and Giα phosphorylation. However, pretreatment with CZI, a PKC-ζ inhibitor, markedly, but not completely, inhibited U46619 induced increase in NADPH oxidase activity, PKC-α activity, Giα phosphorylation and also significantly eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity. Pretreatment with Go6976 inhibited U46619 induced increase in Giα phosphorylation, but not PKC-ζ activity and NADPH oxidase activity. Pretreatment with pertussis toxin eliminated U46619 caused attenuation of isoproterenol stimulated adenyl cyclase activity without any discernible change in PKC-ζ, NADPH oxidase and PKC-α activities. Transfection of the cells with Tp, PKC-ζ and PKC-α siRNA duplexes corroborate the findings observed with their respective pharmacological inhibitors on the responses produced by U46619. Taken together, we suggest involvement of PKC-ζ in U46619 caused attenuation of isoproterenol stimulated ß-adrenergic response, which is regulated by NADPH oxidase-PKCα-Giα axis in pulmonary artery smooth muscle cells.


Subject(s)
15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Myocytes, Smooth Muscle/drug effects , NADPH Oxidases/metabolism , Protein Kinase C-alpha/metabolism , Protein Kinase C/metabolism , Receptors, Adrenergic, beta/metabolism , Adenylyl Cyclases/metabolism , Adrenergic beta-Antagonists/pharmacology , Animals , Cattle , Cyclic AMP/biosynthesis , Enzyme Inhibitors/pharmacology , Isoenzymes/metabolism , Isoproterenol/pharmacology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/antagonists & inhibitors , Pulmonary Artery/cytology
19.
Cell Signal ; 25(2): 512-26, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23159577

ABSTRACT

In the context of cross-talk between transmembrane signaling pathways, we studied the loci within the ß-adrenergic receptor/G protein/adenyl cyclase system at which PKC exerts regulatory effects of peroxynitrite (ONOO(-)) on isoproterenol stimulated adenyl cyclase activity in pulmonary artery smooth muscle cells. Treatment of the cells with ONOO(-) stimulated PKC-α activity and that subsequently increased p(38)MAPK phosphorylation. Pretreatment with Go6976 (PKC-α inhibitor) and SB203580 (p(38)MAPK inhibitor) eliminated ONOO(-) caused inhibition on isoproterenol stimulated adenyl cyclase activity. Pretreatment with Go6976, but not SB203580, prevented ONOO(-) induced increase in PKC-α activity. Studies using genetic inhibitors of PKC-α (PKC-α siRNA) and p(38)MAPK (p(38)MAPK siRNA) also corroborated the findings obtained with their pharmacological inhibitors in eliminating the attenuation of ONOO(-) effect on isoproterenol stimulated adenyl cyclase activity. This inhibitory effect of ONOO(-) was found to be eliminated upon pretreatment of the cells with pertussis toxin thereby pointing to a G(i) dependent mechanism. This hypothesis was reinforced by G(i)α phosphorylation as well as by the observation of the loss of the ability of Gpp(NH)p (a measure of G(i) mediated response) to stimulate adenyl cyclase activity upon ONOO(-) treatment to the cells. We suggest the existence of a pertussis toxin sensitive G protein (G(i))-mediated mechanism in isoproterenol stimulated adenyl cyclase activity, which is regulated by PKCα-p(38)MAPK axis dependent phosphorylation of its α-subunit (G(i)α) in the pulmonary artery smooth muscle cells.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Peroxynitrous Acid/pharmacology , Protein Kinase C-alpha/metabolism , Receptors, Adrenergic, beta/chemistry , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Adenylyl Cyclases/metabolism , Animals , Carbazoles/pharmacology , Cattle , Cell Line , Cyclic AMP/metabolism , Isoproterenol/pharmacology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pertussis Toxin/pharmacology , Phosphorylation , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Pulmonary Artery , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Adrenergic, beta/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
20.
J Biochem ; 153(3): 289-302, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266860

ABSTRACT

We sought to evaluate the mechanism(s) associated with pro matrix metalloprotease 2 (proMMP-2) activation in bovine pulmonary artery smooth muscle cells. Preincubation of cells with anti-TNFR1 antibody prevented tumour necrosis factor-α (TNF-α)-induced proMMP-2 activation and increase in membrane type 1 matrix metalloprotease (MT1-MMP) expression as well as inhibition of tissue inhibitor of metalloproteinase 2 (TIMP-2) expression, indicating the role of TNFR1 receptor during TNF-α stimulation. Anti-MT1-MMP antibody abrogated proMMP-2 activation by TNF-α-stimulated cell membrane, suggesting the involvement of MT1-MMP in proMMP-2 activation. Induction of MT1-MMP expression in response to TNF-α occurs via activation of nuclear factor (NF)-κB on inhibitory κB kinase (IKK) activation and subsequently phosphorylation/degradation of IκB-α. Inhibition of protein kinase C (PKC)-α activity by Go6976 and PKC-α siRNA prevented TNF-α-induced IKK activity, IκB-α phosphorylation/degradation and NF-κB activation. Inhibition of PKC-α activity also prevented TNF-α-induced MT1-MMP expression and proMMP-2 activation as well as down regulation of TIMP-2 expression. Inhibition of IκB-α phosphorylation by PS-1145, an IKK selective inhibitor, prevented TNF-α-induced increase in MT1-MMP expression and proMMP-2 activation, which although did not alter inhibition of TIMP-2 expression. Overall, we unravelled a hitherto unknown mechanism of the involvement of PKC-α in proMMP-2 activation and inhibition of TIMP-2 expression by NF-κB-MT1-MMP-dependent and -independent pathway, respectively, during TNF-α stimulation in pulmonary artery smooth muscle cells.


Subject(s)
Enzyme Precursors/metabolism , Gelatinases/metabolism , Matrix Metalloproteinase 14/metabolism , Myocytes, Smooth Muscle/drug effects , NF-kappa B/metabolism , Protein Kinase C-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cattle , Cells, Cultured , Enzyme Activation/drug effects , Gene Expression/drug effects , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Matrix Metalloproteinase 14/genetics , Models, Biological , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , NF-KappaB Inhibitor alpha , Protein Kinase C-alpha/genetics , Pulmonary Artery/cytology , RNA Interference , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL