Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...