Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Am J Physiol Cell Physiol ; 315(3): C367-C379, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29874111

ABSTRACT

Hypertension is a global health problem, and angiotensin I (ANG I)-converting enzyme (ACE) inhibitors are largely used to control this pathology. Recently, it has been shown that ACE can also act as a transducer signal molecule when its inhibitors or substrates bind to it. This new role of ACE could contribute to understanding some of the effects not explained by its catalytic activity only. In this study, we investigated signaling pathway activation in Chinese hamster ovary (CHO) cells stably expressing ACE (CHO-ACE) under different conditions. We also investigated gene modulation after 4 h and 24 h of captopril treatment. Our results demonstrated that CHO-ACE cells when stimulated with ANG I, ramipril, or captopril led to JNK and ERK1/2 phosphorylation. To verify any physiological role at the endogenous level, we made use of primary cultures of mesangial cells from spontaneously hypertensive rats (SHR) and Wistar rats. Our results showed that ERK1/2 activation occurred mainly in primary cultures of mesangial cells from SHR rats upon captopril stimulation, suggesting that this signaling pathway could be differentially regulated during hypertension. Our results also showed that captopril treatment leads to a decrease of cyclooxygenase 2, interleukin-1ß, and ß-arrestin2 and a significant increase of AP2 gene expression levels. Our findings strengthen the fact that, in addition to the blockage of enzymatic activity, ACE inhibitors also trigger signaling pathway activation, and this may contribute to their beneficial effects in the treatment of hypertension and other pathologies.


Subject(s)
Angiotensin I/metabolism , Captopril/pharmacology , Peptidyl-Dipeptidase A/metabolism , Signal Transduction/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , CHO Cells , Cell Line , Cricetulus , Hypertension/drug therapy , Hypertension/metabolism , MAP Kinase Signaling System/drug effects , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Phosphorylation/drug effects , Rats , Rats, Inbred SHR , Rats, Wistar
2.
Sci Rep ; 6: 22078, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26898917

ABSTRACT

Melanoma is a very aggressive tumor that arises from melanocytes. Late stage and widely spread diseases do not respond to standard therapeutic approaches. The kallikrein-kinin system (KKS) participates in biological processes such as vasodilatation, pain and inflammatory response. However, the role of KKS in tumor formation and progression is not completely understood. The role of the host kinin B1 receptor in melanoma development was evaluated using a syngeneic melanoma model. Primary tumors and metastasis were respectively induced by injecting B16F10 melanoma cells, which are derived from C57BL/6 mice, subcutaneously or in the tail vein in wild type C57BL/6 and B1 receptor knockout mice (B1(-/-)). Tumors developed in B1(-/-) mice presented unfavorable prognostic factors such as increased incidence of ulceration, higher levels of IL-10, higher activation of proliferative pathways such as ERK1/2 and Akt, and increased mitotic index. Furthermore, in the metastasis model, B1(-/-) mice developed larger metastatic colonies in the lung and lower CD8(+)immune effector cells when compared with WT animals. Altogether, our results provide evidences that B1(-/-) animals developed primary tumors with multiple features associated with poor prognosis and unfavorable metastatic onset, indicating that the B1 receptor may contribute to improve the host response against melanoma progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Melanoma, Experimental/genetics , Receptor, Bradykinin B1/genetics , Skin Neoplasms/genetics , Animals , Disease Progression , Female , Interleukin-10/genetics , Interleukin-10/metabolism , Kallikrein-Kinin System/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mitotic Index , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Bradykinin B1/deficiency , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
3.
Clin Sci (Lond) ; 127(3): 185-94, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24498923

ABSTRACT

Regulation of muscle mass depends on the balance between synthesis and degradation of proteins, which is under the control of different signalling pathways regulated by hormonal, neural and nutritional stimuli. Such stimuli are altered in several pathologies, including COPD (chronic obstructive pulmonary disease), diabetes, AIDS and cancer (cachexia), as well as in some conditions such as immobilization and aging (sarcopenia), leading to muscle atrophy, which represents a significant contribution to patient morbidity. The KKS (kallikrein-kinin system) is composed of the enzymes kallikreins, which generate active peptides called kinins that activate two G-protein-coupled receptors, namely B1 and B2, which are expressed in a variety of tissues. The local modulation of the KKS may account for its participation in different diseases, such as those of the cardiovascular, renal and central nervous systems, cancer and many inflammatory processes, including pain. Owing to such pleiotropic actions of the KKS by local modulatory events and the probable fine-tuning of associated signalling cascades involved in skeletal muscle catabolic disorders [for example, NF-κB (nuclear factor κB) and PI3K (phosphoinositide 3-kinase)/Akt pathways], we hypothesized that KKS might contribute to the modulation of intracellular responses in atrophying skeletal muscle. Our results show that kinin B1 receptor activation induced a decrease in the diameter of C2C12 myotubes, activation of NF-κB, a decrease in Akt phosphorylation levels, and an increase in the mRNA levels of the ubiquitin E3 ligases atrogin-1 and MuRF-1 (muscle RING-finger protein-1). In vivo, we observed an increase in kinin B1 receptor mRNA levels in an androgen-sensitive model of muscle atrophy. In the same model, inhibition of the kinin B1 receptor with a selective antagonist resulted in an impairment of atrogin-1 and MuRF-1 expression and IκB (inhibitor of NF-κB) phosphorylation. Moreover, knockout of the kinin B1 receptor in mice led to an impairment in MuRF-1 mRNA expression after induction of LA (levator ani) muscle atrophy. In conclusion, using pharmacological and gene-ablation tools, we have obtained evidence that the kinin B1 receptor plays a significant role in the regulation of skeletal muscle proteolysis in the LA muscle atrophy model.


Subject(s)
Bradykinin/analogs & derivatives , Receptor, Bradykinin B2/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Bradykinin/pharmacology , Kinins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy/genetics , RNA, Messenger/metabolism , Receptor, Bradykinin B2/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Up-Regulation
4.
PLoS One ; 8(5): e64453, 2013.
Article in English | MEDLINE | ID: mdl-23691222

ABSTRACT

Melanoma is a very aggressive tumor that does not respond well to standard therapeutic approaches, such as radio- and chemotherapies. Furthermore, acquiring the ability to metastasize in melanoma and many other tumor types is directly related to incurable disease. The B1 kinin receptor participates in a variety of cancer-related pathophysiological events, such as inflammation and angiogenesis. Therefore, we investigated whether this G protein-coupled receptor plays a role in tumor progression. We used a murine melanoma cell line that expresses the kinin B1 receptor and does not express the kinin B2 receptor to investigate the precise contribution of activation of the B1 receptor in tumor progression and correlated events using various in vitro and in vivo approaches. Activation of the kinin B1 receptor in the absence of B2 receptor inhibits cell migration in vitro and decreases tumor formation in vivo. Moreover, tumors formed from cells stimulated with B1-specific agonist showed several features of decreased aggressiveness, such as smaller size and infiltration of inflammatory cells within the tumor area, higher levels of pro-inflammatory cytokines implicated in the host anti-tumor immune response, lower number of cells undergoing mitosis, a poorer vascular network, no signs of invasion of surrounding tissues or metastasis and increased animal survival. Our findings reveal that activation of the kinin B1 receptor has a host protective role during murine melanoma tumor progression, suggesting that the B1 receptor could be a new anti-tumor GPCR and provide new opportunities for therapeutic targeting.


Subject(s)
Melanoma/prevention & control , Neoplasm Metastasis/prevention & control , Receptor, Bradykinin B1/metabolism , Analysis of Variance , Blotting, Western , Cell Line, Tumor , Fluorescent Dyes , Gene Expression Profiling , Humans , Immunohistochemistry , Kisspeptins/metabolism , Melanoma/physiopathology , Receptor, Bradykinin B1/agonists , Tetrazolium Salts , Thiazoles
5.
Biochim Biophys Acta ; 1832(10): 1591-604, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23643711

ABSTRACT

The mitochondrial redox state plays a central role in the link between mitochondrial overloading and insulin resistance. However, the mechanism by which the ROS induce insulin resistance in skeletal muscle cells is not completely understood. We examined the association between mitochondrial function and H2O2 production in insulin resistant cells. Our hypothesis is that the low mitochondrial oxygen consumption leads to elevated ROS production by a mechanism associated with reduced PGC1α transcription and low content of phosphorylated CREB. The cells were transfected with either the encoded sequence for catalase overexpression or the specific siRNA for catalase inhibition. After transfection, myotubes were incubated with palmitic acid (500µM) and the insulin response, as well as mitochondrial function and fatty acid metabolism, was determined. The low mitochondrial oxygen consumption led to elevated ROS production by a mechanism associated with ß-oxidation of fatty acids. Rotenone was observed to reduce the ratio of ROS production. The elevated H2O2 production markedly decreased the PGC1α transcription, an effect that was accompanied by a reduced phosphorylation of Akt and CREB. The catalase transfection prevented the reduction in the phosphorylated level of Akt and upregulated the levels of phosphorylated CREB. The mitochondrial function was elevated and H2O2 production reduced, thus increasing the insulin sensitivity. The catalase overexpression improved mitochondrial respiration protecting the cells from fatty acid-induced, insulin resistance. This effect indicates that control of hydrogen peroxide production regulates the mitochondrial respiration preventing the insulin resistance in skeletal muscle cells by a mechanism associated with CREB phosphorylation and ß-oxidation of fatty acids.


Subject(s)
Catalase/metabolism , Hydrogen Peroxide/metabolism , Insulin Resistance , Mitochondria, Muscle/physiology , Animals , Antioxidants/metabolism , Cells, Cultured , Male , Mitochondria, Muscle/enzymology , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Oxygen Consumption , Palmitic Acid/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar
6.
Biochem Biophys Res Commun ; 434(3): 647-52, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23583236

ABSTRACT

Mechanotransduction enables cells to sense and respond to stimuli, such as strain, pressure and shear stress (SS), critical for maintenance of cardiovascular homeostasis or pathological states. The angiotensin II type 1 receptor (AT1R) was the first G protein-coupled receptor described to display stretch-induced activation in cardiomyocytes independent of its ligand Ang II. Here, we assessed whether SS (15 dynes/cm(2), 10 min), an important mechanical force present in the cardiovascular system, activates AT1R independent of its ligand. SS induced extracellular signal-regulated kinase (ERK) activation, used as a surrogate of AT1R activation, in Chinese hamster ovary cells expressing the AT1R (CHO+AT1) but not in wild type cells (CHO). AT1R dependent SS-induced ERK activation involves Ca(2+) inflow and activation of Gαq since Ca(2+) chelator EGTA or Gαq-specific inhibitor YM-254890 decreased SS-induced ERK activation. On the other hand, the activation of JAK-2 and Src, two intracellular signaling molecules independent of G protein activation, were not differently modulated in the presence of AT1R. Also, ERK activation by SS was observed in CHO cells expressing the mutated AT1R DRY/AAY, which has impaired ability to activate Gαq dependent intracellular signaling. Altogether we provided evidence that SS activates AT1R in the absence of its ligand by both a G protein-dependent and -independent pathways. The biological relevance of these observations deserves to be further investigated since the novel mechanisms described extend the knowledge of the activation of GPCRs independent of its traditional ligand.


Subject(s)
GTP-Binding Proteins/physiology , Receptor, Angiotensin, Type 1/metabolism , Stress, Physiological , Animals , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Polymerase Chain Reaction
7.
Int J Cardiol ; 167(4): 1199-205, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-22483258

ABSTRACT

BACKGROUND: Hyperactivation of the renin-angiotensin system contributes to hypertension-induced upregulation of vascular matrix metalloproteinases (MMPs) and remodeling, especially in the two kidney, one clip (2K1C) hypertension model. We hypothesized that the AT1R antagonist losartan or the renin inhibitor aliskiren, given at doses allowing similar antihypertensive effects, could prevent in vivo vascular MMPs upregulation and remodeling, and collagen/elastin deposition found in 2K1C hypertension by preventing the activation of extracellular signal-regulated kinase 1/2 (ERK 1/2) and transforming growth factor-ß1 (TGF-ß1). We also hypothesized that aliskiren could enhance the effects of losartan. METHODS: 2K1C rats were treated with aliskiren (50mg.kg(-1).day(-1)), or losartan (10mg.kg(-1).day(-1)), or both by gavage during 4 weeks. RESULTS: Aliskiren, losartan, or both drugs exerted similar antihypertensive effects when compared with 2K-1C rats treated with water. Aliskiren reduced plasma renin activity in both sham and 2K-1C rats. Losartan alone or combined with aliskiren, but not aliskiren alone, abolished 2K1C-induced aortic hypertrophy and hyperplasia, and prevented the increases in aortic collagen/elastin content, MMP-2 levels, gelatinolytic activity, and expression of phospho-ERK 1/2 and TGF-ß1. No significant differences were found in the aortic expression of the (pro)renin receptor. CONCLUSIONS: These findings show that although losartan and aliskiren exerted similar antihypertensive effects, only losartan prevented the activation of vascular profibrotic mechanisms and MMP upregulation associated with vascular remodeling in 2K1C hypertension. Our findings also suggest that aliskiren does not enhance the protective effects exerted by losartan.


Subject(s)
Amides/therapeutic use , Antihypertensive Agents/therapeutic use , Aorta/drug effects , Fumarates/therapeutic use , Hypertension/drug therapy , Losartan/therapeutic use , Ventricular Remodeling/drug effects , Amides/pharmacology , Animals , Antihypertensive Agents/pharmacology , Aorta/metabolism , Aorta/pathology , Fumarates/pharmacology , Hypertension/metabolism , Hypertension/pathology , Losartan/pharmacology , Male , Random Allocation , Rats , Rats, Wistar , Treatment Outcome , Ventricular Remodeling/physiology
8.
Regul Pept ; 177(1-3): 27-34, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22561691

ABSTRACT

We recently demonstrated that Angiotensin-(3-4) [Ang-(3-4)], an Ang II-derived dipeptide, overcomes inhibition of plasma membrane Ca(2+)-ATPase promoted by nanomolar concentrations of Ang II in basolateral membranes of renal proximal tubule cells, with involvement of a so far unknown AT(2)R-dependent and NO-independent mechanism. The present study investigates the signaling pathway triggered by Ang-(3-4) that is responsible for counteracting the inhibitory effect of Ang II, and attempts to elucidate the functional interaction of the dipeptide with Ang II at the level of AT(2)R. Stimulation by cholera toxin of G(s)α protein structurally linked to AT(2)R--as revealed by their co-immunoprecipitation--mimicked the effect of Ang-(3-4) on Ca(2+)-ATPase activity. Furthermore, addition of dibutyril-cAMP (db-cAMP) mimicked Ang-(3-4), whereas the specific PKA inhibitor, PKAi(5-24) peptide, suppressed the counter-regulatory effect of Ang-(3-4) and the AT(2)R agonist, CGP42112A. Membrane-associated PKA activity was stimulated by Ang-(3-4) or CGP42112A to comparable levels as db-cAMP, and the Ang-(3-4) effect was abrogated by the AT(2)R antagonist PD123319, whereas the AT(1)R antagonist Losartan had no effect. Ang-(3-4) stimulated PKA-mediated phosphorylation of Ca(2+)-ATPase and activated PKA to comparable levels. Binding assays demonstrated that Ang-(3-4) could not displace (3)H-Ang II from HEK 293T cells expressing AT(2)R, but 10(-10) mol/L Ang-(3-4) resulted in the appearance of a probable higher-affinity site (picomolar range) for Ang II. The results presented herein demonstrate that Ang-(3-4), acting as an allosteric enhancer, suppresses Ang II-mediated inhibition of Ca(2+)-ATPase through an AT(2)R/cAMP/PKA pathway, after inducing conformational changes in AT(2)R that results in generation of higher-affinity sites for Ang II.


Subject(s)
Angiotensin II/pharmacology , Calcium-Transporting ATPases/metabolism , Cyclic AMP/metabolism , Oligopeptides/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Allosteric Regulation , Angiotensin II/antagonists & inhibitors , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Binding, Competitive , Calcium-Transporting ATPases/antagonists & inhibitors , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Enzyme Assays , HEK293 Cells , Humans , Imidazoles/pharmacology , Immunoprecipitation , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/metabolism , Losartan/pharmacology , Phosphorylation , Protein Binding , Protein Conformation/drug effects , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/agonists , Sheep , Signal Transduction , Transfection
9.
Am J Physiol Renal Physiol ; 302(7): F875-83, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22218590

ABSTRACT

ANG II is secreted into the lumens of proximal tubules where it is also synthesized, thus increasing the local concentration of the peptide to levels of potential physiological relevance. In the present work, we studied the effect of ANG II via the luminal membranes of LLC-PK(1) cells on Ca(2+)-ATPase of the sarco(endo)plasmic reticulum (SERCA) and plasma membrane (PMCA). ANG II (at concentrations found in the lumen) stimulated rapid (30 s) and persistent (30 min) SERCA activity by more than 100% and increased Ca(2+) mobilization. Pretreatment with ANG II for 30 min enhanced the ANG II-induced Ca(2+) spark, demonstrating a positively self-sustained stimulus of Ca(2+) mobilization by ANG II. ANG II in the medium facing the luminal side of the cells decreased with time with no formation of metabolites, indicating peptide internalization. ANG II increased heterodimerization of AT(1) and AT(2) receptors by 140%, and either losartan or PD123319 completely blocked the stimulation of SERCA by ANG II. Using the PLC inhibitor U73122, PMA, and calphostin C, it was possible to demonstrate the involvement of a PLC→DAG(PMA)→PKC pathway in the stimulation of SERCA by ANG II with no effect on PMCA. We conclude that ANG II triggers SERCA activation via the luminal membrane, increasing the Ca(2+) stock in the reticulum to ensure a more efficient subsequent mobilization of Ca(2+). This first report on the regulation of SERCA activity by ANG II shows a new mechanism for Ca(2+) homeostasis in renal cells and also for regulation of Ca(2+)-modulated fluid reabsorption in proximal tubules.


Subject(s)
Angiotensin II/metabolism , Kidney Tubules, Proximal/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Calcium/metabolism , Cell Line , Cell Membrane/enzymology , Neprilysin/metabolism , Peptide Hydrolases/metabolism , Peptidyl-Dipeptidase A/metabolism , Protein Multimerization , Signal Transduction , Swine
10.
Mol Vis ; 17: 2228-40, 2011.
Article in English | MEDLINE | ID: mdl-21897745

ABSTRACT

PURPOSE: The apoptosis of retinal neurons plays a critical role in the pathogenesis of diabetic retinopathy (DR), but the molecular mechanisms underlying this phenomenon remain unclear. The purpose of this study was to investigate the cellular localization and the expression of microRNA-29b (miR-29b) and its potential target PKR associated protein X (RAX), an activator of the pro-apoptotic RNA-dependent protein kinase (PKR) signaling pathway, in the retina of normal and diabetic rats. METHODS: Retinas were obtained from normal and diabetic rats within 35 days after streptozotocin (STZ) injection. In silico analysis indicated that RAX is a potential target of miR-29b. The cellular localization of miR-29b and RAX was assessed by in situ hybridization and immunofluorescence, respectively. The expression levels of miR-29b and RAX mRNA were evaluated by quantitative reverse transcription PCR (qRT-PCR), and the expression of RAX protein was evaluated by western blot. A luciferase reporter assay and inhibition of endogenous RAX were performed to confirm whether RAX is a direct target of miR-29b as predicted by the in silico analysis. RESULTS: We found that miR-29b and RAX are localized in the retinal ganglion cells (RGCs) and the cells of the inner nuclear layer (INL) of the retinas from normal and diabetic rats. Thus, the expression of miR-29b and RAX, as assessed in the retina by quantitative RT-PCR, reflects their expression in the RGCs and the cells of the INL. We also revealed that RAX protein is upregulated (more than twofold) at 3, 6, 16, and 22 days and downregulated (70%) at 35 days, whereas miR-29b is upregulated (more than threefold) at 28 and 35 days after STZ injection. We did not confirm the computational prediction that RAX is a direct target of miR-29b. CONCLUSIONS: Our results suggest that RAX expression may be indirectly regulated by miR-29b, and the upregulation of this miRNA at the early stage of STZ-induced diabetes may have a protective effect against the apoptosis of RGCs and cells of the INL by the pro-apoptotic RNA-dependent protein kinase (PKR) signaling pathway.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Retinal Ganglion Cells/metabolism , Retinal Horizontal Cells/metabolism , Signal Transduction/genetics , eIF-2 Kinase/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/genetics , Gene Expression Regulation , Genes, Reporter , Homeodomain Proteins/genetics , In Situ Hybridization , Luciferases/analysis , Male , MicroRNAs/genetics , Rats , Rats, Wistar , Retinal Ganglion Cells/cytology , Retinal Horizontal Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , eIF-2 Kinase/genetics
11.
Eur J Med Chem ; 46(9): 3616-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21665332

ABSTRACT

This study describes the synthesis of a new ruthenium nitrosyl complex with the formula [RuCl(2)NO(BPA)] [BPA = (2-hydroxybenzyl)(2-methylpyridyl)amine ion], which was synthesized and characterized by spectroscopy, cyclic voltammetry, X-ray crystallography, and theoretical calculation data. The biological studies of this complex included in vitro cytotoxic assays, which revealed its activity against two different tumor cell lines (HeLa and Tm5), with efficacy comparable to that of cisplatin, a metal-based drug that is administered in clinical treatment. The in vivo studies showed that [RuCl(2)NO(BPA)]is effective in reducing tumor mass. Also, our results suggest that the mechanism of action of [RuCl(2)NO(BPA)] includes binding to DNA, causing fragmentation of this biological molecule, which leads to apoptosis.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ruthenium Compounds/chemistry , Ruthenium Compounds/pharmacology , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Spectrum Analysis/methods
12.
Hypertension ; 57(5): 965-72, 2011 May.
Article in English | MEDLINE | ID: mdl-21422380

ABSTRACT

Angiotensin (Ang) I-converting enzyme (ACE) is involved in the control of blood pressure by catalyzing the conversion of Ang I into the vasoconstrictor Ang II and degrading the vasodilator peptide bradykinin. Human ACE also functions as a signal transduction molecule, and the binding of ACE substrates or its inhibitors initiates a series of events. In this study, we examined whether Ang II could bind to ACE generating calcium signaling. Chinese hamster ovary cells transfected with an ACE expression vector reveal that Ang II is able to bind with high affinity to ACE in the absence of the Ang II type 1 and type 2 receptors and to activate intracellular signaling pathways, such as inositol 1,4,5-trisphosphate and calcium. These effects could be blocked by the ACE inhibitor, lisinopril. Calcium mobilization was specific for Ang II, because other ACE substrates or products, namely Ang 1-7, bradykinin, bradykinin 1-5, and N-acetyl-seryl-aspartyl-lysyl-proline, did not trigger this signaling pathway. Moreover, in Tm5, a mouse melanoma cell line endogenously expressing ACE but not Ang II type 1 or type 2 receptors, Ang II increased intracellular calcium and reactive oxygen species. In conclusion, we describe for the first time that Ang II can interact with ACE and evoke calcium and other signaling molecules in cells expressing only ACE. These findings uncover a new mechanism of Ang II action and have implications for the understanding of the renin-Ang system.


Subject(s)
Angiotensin II/metabolism , Calcium Signaling/physiology , Peptidyl-Dipeptidase A/metabolism , Analysis of Variance , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , CHO Cells , Calcium Signaling/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Flow Cytometry , Lisinopril/pharmacology , Mice , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
J Cell Physiol ; 225(2): 500-5, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20458750

ABSTRACT

Eukaryotic translation initiation factor 5A (eIF5A) has a unique character: the presence of an unusual amino acid, hypusine, which is formed by post-translational modifications. Even before the identification of hypusination in eIF5A, the correlation between hypusine formation and protein synthesis, shifting cell proliferation rates, had already been observed. Embryogenesis is a complex process in which cellular proliferation and differentiation are intense. In spite of the fact that many studies have described possible functions for eIF5A, its precise role is under investigation, and to date nothing has been reported about its participation in embryonic development. In this study we show that eIF5A is expressed at all mouse embryonic post-implantation stages with increase in eIF5A mRNA and protein expression levels between embryonic days E10.5 and E13.5. Immunohistochemistry revealed the ubiquitous presence of eIF5A in embryonic tissues and organs at E13.5 day. Interestingly, stronger immunoreactivity to eIF5A was observed in the stomodeum, liver, ectoderm, heart, and eye, and the central nervous system; regions which are known to undergo active differentiation at this stage, suggesting a role of eIF5A in differentiation events. Expression analyses of MyoD, a myogenic transcription factor, revealed a significantly higher expression from day E12.5 on, both at the mRNA and the protein levels suggesting a possible correlation to eIF5A. Accordingly, we next evidenced that inhibiting eIF5A hypusination in mouse myoblast C2C12 cells impairs their differentiation into myotubes and decreases MyoD transcript levels. Those results point to a new functional role for eIF5A, relating it to embryogenesis, development, and cell differentiation.


Subject(s)
Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Peptide Initiation Factors/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Mice , Muscle Fibers, Skeletal/cytology , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Peptide Initiation Factors/antagonists & inhibitors , Peptide Initiation Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Eukaryotic Translation Initiation Factor 5A
14.
Regul Pept ; 141(1-3): 159-67, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17320985

ABSTRACT

Earlier studies with Mas protooncogene, a member of the G-protein-coupled receptor family, have proposed this gene to code for a functional AngII receptor, however further results did not confirm this assumption. In this work we investigated the hypothesis that a heterodimeration AT(1)/Mas could result in a functional interaction between both receptors. For this purpose, CHO or COS-7 cells were transfected with the wild-type AT(1) receptor, a non-functional AT(1) receptor double mutant (C18F-K20A) and Mas or with WT/Mas and C18F-K20A/Mas. Cells single-expressing Mas or C18F/K20A did not show any binding for AngII. The co-expression of the wild-type AT(1) receptor and Mas showed a binding profile similar to that observed for the wild-type AT(1) expressed alone. Surprisingly, the co-expression of the double mutant C18F/K20A and Mas evoked a total recovery of the binding affinity for AngII to a level similar to that obtained for the wild-type AT(1). Functional measurements using inositol phosphate and extracellular acidification rate assays also showed a clear recovery of activity for AngII on cells co-expressing the mutant C18F/K20A and Mas. In addition, immunofluorescence analysis localized the AT(1) receptor mainly at the plasma membrane and the mutant C18F-K20A exclusively inside the cells. However, the co-expression of C18F-K20A mutant with the Mas changed the distribution pattern of the mutant, with intense signals at the plasma membrane, comparable to those observed in cells expressing the wild-type AT(1) receptor. These results support the hypothesis that Mas is able to rescue binding and functionality of the defective C18F-K20A mutant by dimerization.


Subject(s)
Mutation , Proto-Oncogenes/genetics , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Angiotensin II/metabolism , Animals , CHO Cells , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cricetinae , Cricetulus , Fluoresceins , Fluorescent Antibody Technique, Direct , Fluorescent Dyes , Indoles , Inhibitory Concentration 50 , Inositol Phosphates/analysis , Inositol Phosphates/metabolism , Models, Chemical , Molecular Sequence Data , Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/chemistry , Receptors, G-Protein-Coupled/genetics , Transfection
15.
Regul Pept ; 140(1-2): 32-6, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17239455

ABSTRACT

Most of the classical physiological effects of the octapeptide angiotensin II (AngII) are produced by activating the AT1 receptor which belongs to the G-protein coupled receptor family (GPCR). Peptidic GPCRs may be functionally divided in three regions: (i) extracellular domains involved in ligand binding; (ii) intracellular domains implicated in agonist-induced coupling to G protein and (iii) seven transmembrane domains (TM) involved in signal transduction. The TM regions of such receptors have peculiar characteristics such as the presence of proline residues. In this project we aimed to investigate the participation of two highly conserved proline residues (Pro82 and Pro162), located in TM II and TM IV, respectively, in AT1 receptor signal transduction. Both mutations did not cause major alterations in AngII affinity. Functional assays indicated that the P162A mutant did not influence the signal transduction. On the other hand, a potent deleterious effect of P82A mutation on signal transduction was observed. We believe that the Pro82 residue is crucial to signal transduction, although it is not possible to say yet if this is due to a direct participation or if due to a structural rearrangement of TM II. In this last hypothesis, the removal of proline residue might be correlated to a removal of a kink, which in turn can be involved in the correct positioning of residues involved in signal transduction.


Subject(s)
Proline/genetics , Receptor, Angiotensin, Type 1/genetics , Signal Transduction/genetics , Amino Acid Sequence , Angiotensin II/metabolism , Animals , Binding, Competitive , COS Cells , Chlorocebus aethiops , Computer Simulation , Models, Biological , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed/methods , Mutation , Proline/chemistry , Protein Binding , Rats , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL