Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Nitric Oxide ; 148: 1-12, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38636582

ABSTRACT

Epidemiological studies show a strong correlation between diabetes and the increased risk of developing different cancers, including melanoma. In the present study, we investigated the impact of a streptozotocin (STZ)-induced hyperglycemic environment on B16F10-Nex2 murine melanoma development. Hyperglycemic male C57Bl/6 mice showed increased subcutaneous tumor development, partially inhibited by metformin. Tumors showed increased infiltrating macrophages, and augmented IL-10 and nitric oxide (NO) concentrations. In vivo neutralization of IL-10, NO synthase inhibition, and depletion of macrophages reduced tumor development. STZ-treated TLR4 KO animals showed delayed tumor development; the transfer of hyperglycemic C57Bl/6 macrophages to TLR4 KO reversed this effect. Increased concentrations of IL-10 present in tumor homogenates of hyperglycemic mice induced a higher number of pre-angiogenic structures in vitro, and B16F10-Nex2 cells incubated with different glucose concentrations in vitro produced increased levels of IL-10. In summary, our findings show that a hyperglycemic environment stimulates murine melanoma B16F10-Nex2 primary tumor growth, and this effect is dependent on tumor cell stimulation, increased numbers of macrophages, and augmented IL-10 and NO concentrations. These findings show the involvement of tumor cells and other components of the tumor microenvironment in the development of subcutaneous melanoma under hyperglycemic conditions, defining novel targets for melanoma control in diabetic patients.


Subject(s)
Hyperglycemia , Interleukin-10 , Macrophages , Melanoma, Experimental , Mice, Inbred C57BL , Nitric Oxide , Animals , Interleukin-10/metabolism , Nitric Oxide/metabolism , Male , Hyperglycemia/metabolism , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Macrophages/metabolism , Macrophages/drug effects , Mice , Mice, Knockout , Cell Line, Tumor
2.
Nitric Oxide ; 132: 15-26, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36736618

ABSTRACT

Spatial confinement and temporal regulation of signaling by nitric oxide (NO) and reactive oxygen species (ROS) occurs in cancer cells. Signaling mediated by NO and ROS was investigated in two sub clones of the murine melanoma B16F10-Nex2 cell line, Nex10C and Nex8H treated or not with bradykinin (BK). The sub clone Nex10C, similar to primary site cells, has a low capacity for colonizing the lungs, whereas the sub clone Nex8H, similar to metastatic cells, corresponds to a highly invasive melanoma. BK-treated Nex10C cells exhibited a transient increase in NO and an inhibition in basal O2- levels. Inhibition of endogenous NO production by l-NAME resulted in detectable levels of O2-. l-NAME promoted Rac1 activation and enhanced Rac1-PI3K association. l-NAME in the absence of BK resulted in Nex10C cell migration and invasion, suggesting that NO is a negative regulator of O2- mediated cell migration and cell invasion. BK-treated Nex8H cells sustained endogenous NO production through the activation of NOS3. NO activated Rac1 and promoted Rac1-PI3K association. NO stimulated cell migration and cell invasion through a signaling axis involving Ras, Rac1 and PI3K. In conclusion, a role for O2- and NO as positive regulators of Rac1-PI3K signaling associated with cell migration and cell invasion is proposed respectively for Nex10C and Nex8H murine melanoma cells.


Subject(s)
Bradykinin , Melanoma , Mice , Animals , Bradykinin/pharmacology , Bradykinin/metabolism , Superoxides , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Cell Movement
3.
Cell Biol Int ; 46(1): 158-169, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34719858

ABSTRACT

Low levels of nitric oxide (NO) produced by constitutively expressed inducible NO synthase (NOS2) in tumor cells may be an important factor in their development. NOS2 expression is associated with high mortality rates for various cancers. Alternative splicing of NOS2 down-regulates its enzymatic activity, resulting in decreased intracellular NO concentrations. Specific probes to detect alternative splicing of NOS2 were used in two isogenic human colon cancer cell lines derived either from the primary tumor (SW480) or from a lymph node metastasis (SW620). Splicing variant of NOS2 S3, lacking exons 9, 10, and 11, was overexpressed in SW480 cells. NOS2 S3 was silenced in SW480 cells. Flow-cytometry analysis was used to estimate the intracellular NO levels and to analyze the cell cycle of the studied cell lines. Western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR) were used to determine apoptosis and autophagy markers. SW480 and SW620 cells expressed NOS2 S3. Overexpression of the NOS2 S3 in SW480 cells downregulated intracellular NO levels. SW480 cells with knocked down NOS2 S3 (referred to as S3C9 cells) had higher intracellular levels of NO compared to the wild-type SW480 cells under serum restriction. Higher NO levels resulted in the loss of viability of S3C9 cells, which was associated with autophagy. Induction of autophagy by elevated intracellular NO levels in S3C9 cells under serum restriction, suggests that autophagy operates as a cytotoxic response to nitrosative stress. The expression of NOS2 S3 plays an important role in regulating intracellular NO production and maintaining viability in SW480 cells under serum restriction. These findings may prove significant in the design of NOS2/NO-based therapies for colon cancer.


Subject(s)
Adenocarcinoma/enzymology , Autophagy , Colonic Neoplasms/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Nitrosative Stress , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Nitric Oxide Synthase Type II/genetics , Protein Isoforms , Signal Transduction
4.
Nitric Oxide ; 93: 78-89, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31539562

ABSTRACT

Human immunodeficiency virus (HIV) infections are typically accompanied by high levels of secreted inflammatory cytokines and generation of high levels of reactive oxygen species (ROS). To elucidate how HIV-1 alters the cellular redox environment during viral replication, we used human HIV-1 infected CD4+T lymphocytes and uninfected cells as controls. ROS and nitric oxide (NO) generation, antioxidant enzyme activity, protein phosphorylation, and viral and proviral loads were measured at different times (2-36 h post-infection) in the presence and absence of the NO donor S-nitroso-N-acetylpenicillamine (SNAP). HIV-1 infection increased ROS generation and decreased intracellular NO content. Upon infection, we observed increases in copper/zinc superoxide dismutase (SOD1) and glutathione peroxidase (GPx) activities, and a marked decrease in glutathione (GSH) concentration. Exposure of HIV-1 infected CD4+T lymphocytes to SNAP resulted in an increasingly oxidizing intracellular environment, associated with tyrosine nitration and SOD1 inhibition. In addition, SNAP treatment promoted phosphorylation and activation of the host's signaling proteins, PKC, Src kinase and Akt. Inhibition of PKC leads to inhibition of Src kinase strongly suggesting that PKC is the upstream element in this signaling cascade. Changes in the intracellular redox environment after SNAP treatment had an effect on HIV-1 replication as reflected by increases in proviral and viral loads. In the absence or presence of SNAP, we observed a decrease in viral load in infected CD4+T lymphocytes pre-incubated with the PKC inhibitor GF109203X. In conclusion, oxidative/nitrosative stress conditions derived from exposure of HIV-1-infected CD4+T lymphocytes to an exogenous NO source trigger a signaling cascade involving PKC, Src kinase and Akt. Activation of this signaling cascade appears to be critical to the establishment of HIV-1 infection.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , HIV-1/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Virus Replication/physiology , HIV Infections , Humans , Nitric Oxide Donors/pharmacology , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , src-Family Kinases/metabolism
5.
Nitric Oxide ; 89: 1-13, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31009708

ABSTRACT

Cancer development is closely related to chronic inflammation, which is associated with identifiable markers of tumor progression, such as uncontrolled cell proliferation, angiogenesis, genomic instability, chemotherapeutic resistance, and metastases. Redox processes mediated by reactive oxygen species (ROS) and nitric oxide (NO) within the inflammatory tumor microenvironment play an essential role in directly influencing intercellular and intracellular signaling. These reactive species originating in the cancer cell or its microenvironment, mediate the epithelial-mesenchymal transition (EMT) and the mesenchymal-epithelial transition (MET). However, intracellular interactions between NO and ROS must be controlled to prevent cell death. Melanoma, breast, and colon cancer cells have developed a mechanism to survive and adapt to oxidative and nitrosative stress. The mechanism involves a spatial-temporal fine adjustment of the intracellular concentrations of NO and ROS, thereby guaranteeing the successful development of cancer cells. Physiological concentrations of NO and supra physiological concentrations of ROS are prevalent in cancer cells at the primary site. The situation reverses in cancer cells undergoing the EMT prior to being released into the blood stream. Intracellular supra physiological concentrations of NO found in circulating cancer cells endow them with anoikis resistance. When the anoikis-resistant cancer cells arrive at a metastatic site they undergo the MET. Endogenous supra physiological concentrations of ROS and physiological NO concentrations are prevalent in these cells. Understanding tumor progression from the perspective of redox signaling permits the characterization of new markers and approaches to therapy. The synthesis and use of compounds with the capacity of modifying intracellular concentrations of NO and ROS may prove effective in disrupting a redox homeostasis operative in cancer cells.


Subject(s)
Breast Neoplasms/physiopathology , Colonic Neoplasms/physiopathology , Melanoma/physiopathology , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/physiology , Humans , Melanoma/drug therapy , Nitric Oxide Donors/therapeutic use , Tumor Microenvironment/physiology
6.
Free Radic Res ; 52(5): 592-604, 2018 May.
Article in English | MEDLINE | ID: mdl-29651879

ABSTRACT

Tumour progression involves the establishment of tumour metastases at distant sites. Resistance to anoikis, a form of cell death that occurs when cells lose contact with the extracellular matrix and with neighbouring cells, is essential for metastases. NO has been associated with anoikis. NO treated HeLa cells and murine melanoma cells in suspension triggered a nitric oxide (NO)-Src kinase signalling circuitry that enabled resistance to anoikis. Two NO donors, sodium nitroprusside (SNP) (500 µM) and DETANO (125 µM), protected against cell death derived from detachment of a growth permissive surface (experimental anoikis). Under conditions of NO-mediated Src activation the following were observed: (a) down-regulation of the pro-apoptotic proteins Bim and cleaved caspase-3 and the cell surface protein, E-cadherin, (b) up-regulation of caveolin-1, and (c) the dissociation of cell aggregates formed when cells are detached from a growth permissive surface. Efficiency of reattachment of tumour cells in suspension and treated with different concentrations of an NO donor, was dependent on the NO concentration. These findings indicate that NO-activated Src kinase triggers a signalling circuitry that provides resistance to anoikis, and allows for metastases.


Subject(s)
Anoikis/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/pharmacology , Nitroprusside/pharmacology , Nitroso Compounds/pharmacology , src-Family Kinases/genetics , Animals , Anoikis/genetics , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Enzyme Activation/drug effects , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Nitric Oxide/chemistry , Nitric Oxide Donors/chemistry , Nitroprusside/chemistry , Nitroso Compounds/chemistry , Signal Transduction , Tumor Cells, Cultured , src-Family Kinases/metabolism
7.
J Cell Physiol ; 233(4): 3176-3194, 2018 04.
Article in English | MEDLINE | ID: mdl-28833096

ABSTRACT

We investigated the role of glycosaminoglycans (GAGs) in the regulation of endothelial nitric oxide synthase (eNOS) activity in wild-type CHO-K1 cells and in xylosyltransferase-deficient CHO-745 cells. GAGs inhibit the integrin/FAK/PI3K/AKT signaling pathway in CHO-K1 cells, decreasing the phosphorylation of eNOS at Ser1177. Furthermore, in CHO-K1 cells, eNOS and PKCα are localized at sphingolipid- and cholesterol-rich domains in the plasma membrane called caveolae. At caveolae, PKCα activation stimulates the phosphorylation of eNOS on Thr495, resulting in further inhibition of NO production in these cells. In our data, CHO-745 cells generate approximately 12-fold more NO than CHO-K1 cells. Increased NO production in CHO-745 cells promotes higher rates of protein S-nitrosylation and protein tyrosine nitration. Regarding reactive oxygen species (ROS) production, CHO-745 cells show lower basal levels of superoxide (O2- ) than CHO-K1 cells. In addition, CHO-745 cells express higher levels of GPx, Trx1, and catalase than CHO-K1 cells, suggesting that CHO-745 cells are in a constitutive nitrosative/oxidative stress condition. Accordingly, we showed that CHO-745 cells are more sensitive to oxidant-induced cell death than CHO-K1 cells. The high concentration of NO and reactive oxygen species generated by CHO-745 cells can induce simultaneous mitochondrial biogenesis and antioxidant gene expression. These observations led us to propose that GAGs are part of a regulatory mechanism that participates in eNOS activation and consequently regulates nitrosative/oxidative stress in CHO cells.


Subject(s)
Heparan Sulfate Proteoglycans/deficiency , Intracellular Space/metabolism , Nitric Oxide/biosynthesis , Up-Regulation , Animals , CHO Cells , Cricetinae , Cricetulus , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Nitric Oxide Synthase Type III/metabolism , Oligopeptides/metabolism , Organelle Biogenesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Subcellular Fractions/metabolism
8.
Biomed J ; 40(4): 189-199, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28918907

ABSTRACT

Accumulating mutations may drive cells into the acquisition of abnormal phenotypes that are characteristic of cancer cells. Cancer cells feature profound alterations in proliferation programs that result in a new population of cells that overrides normal tissue construction and maintenance programs. To achieve this goal, cancer cells are endowed with up regulated survival signaling pathways. They also must counteract the cytotoxic effects of high levels of nitric oxide (NO) and of reactive oxygen species (ROS), which are by products of cancer cell growth. Accumulating experimental evidence associates cancer cell survival with their capacity to up-regulate antioxidant systems. Elevated expression of the antioxidant protein thioredoxin-1 (Trx1) has been correlated with cancer development. Trx1 has been characterized as a multifunctional protein, playing different roles in different cell compartments. Trx1 migrates to the nucleus in cells exposed to nitrosative/oxidative stress conditions. Trx1 nuclear migration has been related to the activation of transcription factors associated with cell survival and cell proliferation. There is a direct association between the p21Ras-ERK1/2 MAP Kinases survival signaling pathway and Trx1 nuclear migration under nitrosative stress. The expression of the cytoplasmic protein, the thioredoxin-interacting protein (Txnip), determines the change in Trx1 cellular compartmentalization. The anti-apoptotic actions of Trx1 and its denitrosylase activity occur in the cytoplasm and serve as important regulators of cell survival. Within this context, this review focuses on the participation of Trx1 in cells under nitrosative/oxidative stress in survival signaling pathways associated with cancer development.


Subject(s)
Antioxidants/pharmacology , Cell Survival/drug effects , Neoplasms/drug therapy , Oxidative Stress/drug effects , Thioredoxins/pharmacology , Humans , Oxidation-Reduction/drug effects , Oxidative Stress/physiology
10.
Peptides ; 85: 1-15, 2016 11.
Article in English | MEDLINE | ID: mdl-27575453

ABSTRACT

The present work aims at investigating the mechanism of action of the Rb9 peptide, which contains the VHCDR 3 sequence of anti-sodium-dependent phosphate transport protein 2B (NaPi2B) monoclonal antibody RebMab200 and displayed antitumor properties. Short peptides corresponding to the hypervariable complementarity-determining regions (CDRs) of immunoglobulins have been associated with antimicrobial, antiviral, immunomodulatory and antitumor activities regardless of the specificity of the antibody. We have shown that the CDR derived peptide Rb9 induced substrate hyperadherence, inhibition of cell migration and matrix invasion in melanoma and other tumor cell lines. Rb9 also inhibited metastasis of murine melanoma in a syngeneic mouse model. We found that Rb9 binds to and interferes with Hsp90 chaperone activity causing attenuation of FAK-Src signaling and downregulation of active Rac1 in B16F10-Nex2 melanoma cells. The peptide also bound to an adhesion G-protein coupled receptor, triggering a concentration-dependent synthesis of cAMP and activation of PKA and VASP signaling as well as IP-3 dependent Ca2+ release. Hsp90 is highly expressed on the cell surface of melanoma cells, and synthetic agents that target Hsp90 are promising cancer therapeutic drugs. Based on their remarkable antitumor effects, the CDR-H3-derived peptides from RebMab200, and particularly the highly soluble and stable Rb9, are novel candidates to be further studied as potential antitumor drugs, selectively acting on cancer cell motility and invasion.


Subject(s)
Complementarity Determining Regions/genetics , HSP90 Heat-Shock Proteins/genetics , Melanoma, Experimental/drug therapy , Peptides/genetics , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Movement/genetics , Complementarity Determining Regions/immunology , HSP90 Heat-Shock Proteins/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Neoplasm Invasiveness/genetics , Neuropeptides/genetics , Peptides/administration & dosage , Peptides/immunology , Receptors, G-Protein-Coupled/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/immunology , rac1 GTP-Binding Protein/genetics
11.
Cell Biol Int ; 40(7): 796-802, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27109745

ABSTRACT

Much attention has been drawn to the pro-inflammatory condition that accompanies aging. This study compared parameters from non-stimulated neutrophils, obtained from young (18-30 years old [y.o.]) and elderly (65-80 y.o.) human volunteers. Measured as an inflammatory marker, plasmatic concentration of hs-CRP was found higher in elderly individuals. Non-stimulated neutrophil production of ROS and NO was, respectively, 38 and 29% higher for the aged group. From the adhesion molecules evaluated, only CD11b expression was elevated in neutrophils from the aged group, whereas no differences were found for CD11a, CD18, or CD62. A 69% higher non-stimulated in vitro neutrophil/endothelial cell adhesion was observed for neutrophils isolated from elderly donors. Our results suggest that with aging, neutrophils may be constitutively producing more reactive species in closer proximity to endothelial cells of vessel walls, which may both contribute to vascular damage and reflect a neutrophil intracellular disrupted redox balance, altering neutrophil function in aging.


Subject(s)
Endothelial Cells/cytology , Neutrophils/cytology , Adult , Age Factors , Aged , Aging/physiology , Antigens, CD/metabolism , C-Reactive Protein/metabolism , CD11b Antigen/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , Male , Neutrophils/metabolism , Reactive Oxygen Species/metabolism
12.
Biomed J ; 38(5): 380-8, 2015.
Article in English | MEDLINE | ID: mdl-26068128

ABSTRACT

Cancer is a worldwide health problem leading to a high incidence of morbidity and mortality. Malignant transformation can occur by expression of oncogenes, over-expression and deregulated activation of proto-oncogenes, and inactivation of tumor suppressor genes. These cellular actions occur through stimulation of oncogenic signaling pathways. Nitric oxide (NO) can induce genetic changes in cells and its intracellular generation can lead to tumor formation and progression. It can also promote anti-tumor activities. The pro- and anti-tumor activities of NO are dependent on its intracellular concentration, cell compartmentalization, and cell sensitivity. NO affects a number of oncogenic signaling pathways. This review focuses on two oncogenic signaling pathways: NO-EGFR-Src-FAK and NO-Ras-EGFR-ERK1/2 MAP kinases. In these pathways, low to intermediate concentrations of NO/S-nitrosothiols (RSNOs) stimulate oncogenic signaling, while high concentrations of NO/RSNO stimulate anti-oncogenic signaling. Increasing knowledge on pro- and anti-tumorigenic activities of NO and related reactive species such as RSNOs has fostered the research and synthesis of novel NO-based chemotherapeutic agents. RSNOs, effective as NO donors and trans-nitrosylating agents under appropriate conditions, may operate as potential chemotherapeutic agents.


Subject(s)
Neoplasms/metabolism , Nitric Oxide/metabolism , Protein S/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , Humans , Neoplasms/therapy , Phosphorylation
13.
Nitric Oxide ; 47: 40-51, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25819133

ABSTRACT

The small GTP-binding proteins Ras and Rac1 are molecular switches exchanging GDP for GTP and converting external signals in response to a variety of stimuli. Ras and Rac1 play an important role in cell proliferation, cell differentiation, and cell migration. Rac1 is directly involved in the reorganization and changes in the cytoskeleton during cell motility. Nitric oxide (NO) stimulates the Ras - ERK1/2 MAP kinases signaling pathway and is involved in the interaction between Ras and the phosphatidyl-inositol-3 Kinase (PI3K) signaling pathway and cell migration. This study utilizes bradykinin (BK), which promotes endogenous production of NO, in an investigation of the role of NO in the activation of Rac1 in rabbit aortic endothelial cells (RAEC). NO-derived from BK stimulation of RAEC and incubation of the cells with the s-nitrosothiol S-nitrosoglutathione (GSNO) activated Rac1. NO-derived from BK stimulation promoted RAEC migration over a period of 12 h. The use of RAEC permanently transfected with the dominant negative mutant of Ras (Ras(N17)) or with the non-nitrosatable mutant of Ras (Ras(C118S)); and the use of specific inhibitors of: Ras, PI3K, and Rac1 resulted in inhibition of NO-mediated Rac1 activation. BK-stimulated s-nitrosylation of Ras in RAEC mediates Rac1 activation and cell migration. Inhibition of NO-mediated Rac1 activation resulted in inhibition of endothelial cell migration. In conclusion, the NO indirect activation of Rac1 involves the direct participation of Ras and PI3K in the migration of endothelial cells stimulated with BK.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/drug effects , Nitric Oxide/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , ras Proteins/metabolism , Bradykinin/pharmacology , Endothelial Cells/metabolism , Humans , Nitric Oxide/biosynthesis
14.
Arch Biochem Biophys ; 558: 14-27, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24960080

ABSTRACT

Nitric oxide (NO) is involved in angiogenesis and stimulates the EGF-R signaling pathway. Stimulation of different endothelial cell lines with bradykinin (BK) activates the endothelial NO synthase (eNOS) and promotes EGF-R tyrosine phosphorylation. Increase in NO production correlated with enhanced phosphorylation of tyrosine residues and S-nitrosylation of the EGF-R. NO-mediated stimulatory effects on tyrosine phosphorylation of the EGF-R, where cGMP independent. Inhibition of soluble guanylyl cyclase followed by BK stimulation of human umbilical vein endothelial cells (HUVECs) did not change tyrosine phosphorylation levels of EGF-R. BK-stimulation of HUVEC promoted S-nitrosylation of the phosphatase SHP-1 and of p21Ras. Phosphorylation and activation of the ERK1/2 MAP kinases mediated by BK was dependent on the activation of the B2 receptor, of the EGF-R, and of p21 Ras. Inhibition of BK-stimulated S-nitrosylation prevented the activation of the ERK1/2 MAP kinases. Furthermore, activated ERK1/2 MAP kinases inhibited internalization of EGF-R by phosphorylating specific Thr residues of its cytoplasmic domain. BK-induced proliferation of endothelial cells was partially inhibited by the NOS inhibitor (L-NAME) and by the MEK inhibitor (PD98059). BK stimulated the expression of vascular endothelial growth factor (VEGF). VEGF expression was dependent on the activation of the EGF-R, the B2 receptor, p21Ras, and on NO generation. A Matrigel®-based in vitro assay for angiogenesis showed that BK induced the formation of capillary-like structures in HUVEC, but not in those cells expressing a mutant of the EGF-R lacking tyrosine kinase activity. Additionally, pre-treatment of BK-stimulated HUVEC with L-NAME, PD98059, and with SU5416, a specific inhibitor of VEGFR resulted in inhibition of in vitro angiogenesis. Our findings indicate that BK-mediated angiogenesis in endothelial cells involves the induction of the expression of VEGF associated with the activation of the NO/EGF-R/p21Ras/ERK1/2 MAP kinases signaling pathway.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Bradykinin/pharmacology , ErbB Receptors/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Nitric Oxide/metabolism , Signal Transduction/drug effects , Animals , Cell Proliferation/drug effects , ErbB Receptors/genetics , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nitric Oxide/biosynthesis , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Rabbits , S-Nitrosothiols/metabolism , Tyrosine/metabolism
15.
Antioxid Redox Signal ; 18(3): 221-38, 2013 Jan 20.
Article in English | MEDLINE | ID: mdl-22894707

ABSTRACT

AIMS: S-nitrosylation of Cys118 is a redox-based mechanism for Ras activation mediated by nitric oxide (NO) at the plasma membrane. RESULTS: Ras signaling pathway stimulation by 50 and/or 100 µM of S-nitrosoglutathione (GSNO) causes proliferation of HeLa cells. Proliferation was not observed in HeLa cells overexpressing non-nitrosatable H-Ras(C118S). HeLa cells overexpressing H-Ras(wt) containing the spatiotemporal probe green fluorescent protein (GFP) fused to the Ras-binding domain of Raf-1 (GFP-RBD) incubated with 100 µM GSNO stimulated a rapid and transient redistribution of GFP-RBD to the plasma membrane, followed by a delayed and sustained recruitment to the Golgi. No activation of H-Ras at the plasma membrane occurred in cells overexpressing H-Ras(C118S), contrasting with a robust and sustained activation of the GTPase at the Golgi. Inhibition of Src kinase prevented cell proliferation and activation of H-Ras by GSNO at the Golgi. Human umbilical vein endothelial cells (HUVECs) stimulated with bradykinin to generate NO were used to differentiate cell proliferation and Ras activation at the plasma membrane versus Golgi. In this model, Src kinase was not involved in cell proliferation, whereas Ras activation proceeded only at the plasma membrane, indicating that HUVEC proliferation induced by NO resulted only from stimulation of Ras. INNOVATION: The present work is the first to demonstrate that NO-mediated activation of Ras in different subcellular compartments regulates different downstream signaling pathways. CONCLUSION: S-nitrosylation of H-Ras at Cys(118) and the activation of Src kinase are spatiotemporally linked events of the S-nitrosothiol-mediated signaling pathway that occurs at the plasma membrane and at the Golgi. The nonparticipation of Src kinase and the localized production of NO by endothelial NO synthase at the plasma membrane limited NO-mediated Ras activation to the plasma membrane.


Subject(s)
Cell Proliferation , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , S-Nitrosoglutathione/pharmacology , Animals , Bradykinin/pharmacology , COS Cells , Calcium Signaling , Cell Membrane/enzymology , Chlorocebus aethiops , Cysteine/analogs & derivatives , Cysteine/metabolism , Enzyme Activation , Golgi Apparatus/enzymology , HeLa Cells , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Nitric Oxide/physiology , Oxidation-Reduction , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/metabolism , Protein Processing, Post-Translational , S-Nitrosothiols/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
16.
Drug Chem Toxicol ; 35(3): 324-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22288377

ABSTRACT

Expression of cytochrome P4502E1 (CYP2E1) is very much influenced by nutritional factors, especially carbohydrate consumption, and various results concerning the expression of CYP2E1 were obtained with a low-carbohydrate diet. This study describes the effects of ethanol treatment on CYP2E1 levels and its relationship with oxidative stress using a balanced standard diet to avoid low or high carbohydrate consumption. Rats were fed for 1, 2, 3, or 4 weeks a commercial diet plus an ethanol-sucrose solution. The results have shown that ethanol administration was associated with CYP2E1 induction and stabilization without related oxidative stress. Our findings suggest that experimental models with a low-carbohydrate/high-fat diet produce some undesirable CYP2E1 changes that are not present when a balanced standard diet is given.


Subject(s)
Cytochrome P-450 CYP2E1/metabolism , Diet , Enzyme Induction/drug effects , Ethanol/toxicity , Liver/drug effects , Oxidative Stress/drug effects , Alanine Transaminase/blood , Analysis of Variance , Animals , Aspartate Aminotransferases/blood , Ethanol/administration & dosage , Glutathione/metabolism , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Statistics, Nonparametric
17.
J Cell Physiol ; 227(6): 2740-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21898414

ABSTRACT

Endothelial cells (ECs) are a source of physiologically important molecules that are synthesized and released to the blood and/or to the subendothelial extracellular matrix such as a heparan sulfate proteoglycan (HSPG) with antithrombotic properties. Previously, we have shown that heparin stimulates the synthesis and modifies the sulfation pattern of this HSPG. Here the molecular mechanisms involved in the up-regulation of HSPG synthesis by heparin in endothelial cells were decoded. The cells were stimulated with heparin and the expression of HSPG and intracellular pathways were evaluated by a combination of methods involving confocal microscopy, flow cytometry, Western blotting analyses, and [(35) S]-sulfate metabolically labeling of the cells. We observed that the up-regulation of HSPG synthesis evoked by heparin is dependent on the interaction of heparin with integrin since RGD peptide abolishes the effect. The activation of integrin leads to tyrosine-phosphorylation of focal adhesion-associated proteins such as FAK, Src, and paxillin. In addition, heparin induces ERK1/2 phosphorylation and inhibitors of Ras and MEK decreased heparin-dependent HSPG synthesis. Moreover, heparin also induced intracellular Ca(2+) release, PLCγ1 (phospholipase Cγ1) and CaMKII (calcium calmodulin kinase II) activation, as well as an increase in nitric oxide (NO) production. Finally, an intracellular Ca(2+) chelator, Ca(2+) signaling inhibitors, and an endothelial NO synthase inhibitor were all able to abolish the effect in heparan sulfate synthesis. In conclusion, the heparin-induced up-regulation of HSPG expression is associated with the phosphorylation of focal adhesion proteins and Ras/Raf/MEK/ERK MAP and Ca(2+) /NO pathways.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation/physiology , Heparin/metabolism , Heparitin Sulfate/metabolism , Integrins/metabolism , Animals , Anticoagulants/metabolism , Anticoagulants/pharmacology , Blotting, Western , Calcium/metabolism , Calcium Signaling , Cell Adhesion , Fibronectins/chemistry , Fibronectins/metabolism , Flow Cytometry , Heparin/pharmacology , Heparitin Sulfate/genetics , Microscopy, Confocal , Nitric Oxide , Phosphorylation , Protein Binding , Rabbits , Up-Regulation
18.
Redox Rep ; 16(1): 27-37, 2011.
Article in English | MEDLINE | ID: mdl-21605496

ABSTRACT

We investigated the role of protein tyrosine phosphatase-alpha (PTPα) expression in the cell death profile of the A431 human carcinoma cell line that was induced by cytotoxic concentrations of the nitric oxide (NO) donors sodium nitroprusside (SNP) and 3,3-bis-(aminoethyl)-1-hydroxy-2-oxo-1-triazene (NOC-18). Both NO donors promoted extensive cell detachment in A431 parental cells as compared to the detachment observed for A431 cells that ectopically expressed PTPα (A431 (A27B(PTPα)) cells). The NO-induced cell death characteristics for both cell lines were examined. After incubation for 10 hours with 2.0 mM SNP, attached or detached A431 cells underwent apoptosis. Cells were highly positive for Annexin-V, featured increased cleavage of procaspase-8, activation of downstream caspase-3, and activation of poly-ADP-ribose polymerase 1 (PARP-1). In contrast, exposure of A431 (A27B(PTPα)) cells to 2.0 mM SNP produced an increase in the release of lactate dehydrogenase and enhanced incorporation of propidium iodide. In addition, A431 (A27B(PTPα)) cells showed partial inhibition of the activities of caspase-8, caspase-3, and PARP-1 upon detachment and cell death induced by SNP treatment. Results indicate that necrotic cell damage was induced, characterized by cellular swelling and lysis. We conclude from these results that PTPα regulates the A431 tumor cell death profile mediated by NO donors. Expression of PTPα or its absence may determine the occurrence of NO-induced cell death with necrotic or apoptotic features, respectively.


Subject(s)
Apoptosis , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Nitroso Compounds/pharmacology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Caspases/drug effects , Cell Line, Tumor , Cell Membrane Permeability , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Humans , L-Lactate Dehydrogenase/drug effects , Nitric Oxide/metabolism , Phosphatidylserines/analysis , Poly(ADP-ribose) Polymerases/drug effects , Propidium/analysis , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Transfection
19.
Free Radic Biol Med ; 49(6): 997-1007, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20542107

ABSTRACT

We have previously shown that ARG2 expression was increased in most malignant thyroid tumors, but absent in benign lesions and normal tissues. Small interfering RNA knockdown was used to investigate the role of ARG2 in a thyroid carcinoma cell line. ARG2 knockdown decreased eNOS expression as well as the expression of eNOS-related genes (p21, Akt1, HIF-1, VEGF, and CAV1). ARG2 silencing changed tumor properties of thyroid cancer cells promoting apoptosis and reduced expression of cell proliferation markers. These results, coupled with enhanced nitric oxide production and elevated reactive oxygen species (ROS) levels, account for the altered intracellular redox environment. Genes related to either production (DUOX1 and NOX4) or catabolism (SODs) of ROS and reactive nitrogen species were negatively modulated by ARG2 knockdown. Additionally, a positive correlation of ARG2 with eNOS and related genes was investigated in thyroid tumors, further substantiating our in vitro findings. Our results suggest that ARG2 and eNOS may work in a coordinated manner and the underlying mechanism might be of major significance for thyroid tumorigenesis and/or tumor progression pathways. Fine modulation of ARG2, eNOS, and related genes may represent a potential source for targeted therapy of several cancer types.


Subject(s)
Arginase/metabolism , Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Nitric Oxide Synthase/metabolism , Thyroid Neoplasms/metabolism , Apoptosis/genetics , Arginase/genetics , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/physiopathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Humans , Nitric Oxide Synthase/genetics , Oxidative Stress/genetics , RNA, Small Interfering/genetics , Signal Transduction/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/physiopathology
20.
Antioxid Redox Signal ; 13(2): 109-25, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20055753

ABSTRACT

The role of NO in regulating the focal adhesion proteins, Src, FAK, p130 Cas, and PTP-alpha, was investigated. Fibroblasts expressing PTP-alpha (PTP-alpha(WT) cells), fibroblasts "knockout" for PTP-alpha (PTP-alpha(-/-) cells), and "rescued" "knockout" fibroblasts (PTP-alpha A5/3 cells) were stimulated with either S-nitroso-N-acetylpenicillamine (SNAP) or fetal bovine serum (FBS). FBS increased inducible NO synthase in both cell lines. Activation of Src mediated either by SNAP or by FBS occurred independent of dephosphorylation of Tyr527 in PTP-alpha(-/-) cells. Both stimuli promoted dephosphorylation of Tyr527 and activation of Src kinase in PTP-alpha(WT) cells. NO-mediated activation of Src kinase affected the activities of FAK and p130Cas and was dependent on the expression of PTP-alpha. Analogous to tyrosine phosphorylation, SNAP and FBS stimulated differential generation of NO and S-nitrosylation of Src kinase in both cell lines. Incubation with SNAP resulted in higher levels of NO and S-nitrosylation of immunoprecipitated Src in PTP-alpha(-/-) cells (oxidizing redox environment) as compared with the levels of NO and S-nitrosylated Src in PTP-alpha(WT) cells (reducing redox environment). SNAP differentially stimulated cell proliferation of both cell lines is dependent on the intracellular redox environment, Src activity, and PTP-alpha expression. This dependence also is observed with FBS-stimulated cell migration.


Subject(s)
Crk-Associated Substrate Protein/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Nitric Oxide/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , src-Family Kinases/metabolism , Animals , Cattle , Cell Movement , Cell Proliferation , Cells, Cultured , Crk-Associated Substrate Protein/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Focal Adhesion Protein-Tyrosine Kinases/genetics , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide Donors/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidation-Reduction , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , S-Nitroso-N-Acetylpenicillamine/metabolism , src-Family Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...