Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Semin Cancer Biol ; 43: 35-41, 2017 04.
Article in English | MEDLINE | ID: mdl-28104391

ABSTRACT

Breast cancer is the leading cause of cancer-related death in women in Europe and North America, and metastasis is the primary cause of fatality in patients with breast cancer. While some breast cancers are quite treatable, the triple-negative breast cancers are more metastatic and resistant to chemotherapy. There is clearly an urgent need for better treatments for this form of the disease. Breast cancer is characterized by genetically complex intra-tumour heterogeneity, particularly within the triple-negative clinical subtype. This complicates treatment options, so the development of specifically targeted chemotherapy for less treatable forms is critical. Dysregulation of pH homeostasis is a common factor in breast tumour cells. This occurs in concert with a metabolic switch to aerobic glycolysis that occurs at the onset of oncogenic transformation. The Na+/H+ exchanger isoform 1 (NHE1) is the major pH regulatory protein involved in the increased proton extrusion of breast cancer cells. Its increased activity results in intracellular alkalinisation and extracellular acidification that drives cancer progression. The acidification of the extracellular tumour microenvironment also contributes to the development of chemotherapy resistance. In this review, we outline the role of H+ as a carcinogenic signal and the role and regulation of NHE1 as a trigger for metastasis. We review recent evidence supporting the use of pharmacological inhibitors of NHE1 as a viable treatment option for triple-negative breast cancer.


Subject(s)
Sodium-Hydrogen Exchanger 1/metabolism , Triple Negative Breast Neoplasms/metabolism , Female , Homeostasis , Humans , Hydrogen-Ion Concentration , Signal Transduction , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/etiology
2.
Cell Signal ; 29: 69-77, 2017 01.
Article in English | MEDLINE | ID: mdl-27751915

ABSTRACT

Mounting evidence supports a major role for the Na+/H+ exchanger NHE1 in cancer progression and metastasis. NHE1 is hyperactive at the onset of oncogenic transformation, resulting in intracellular alkalinization and extracellular microenvironmental acidification. These conditions promote invasion and facilitate metastasis. However, the signal pathways governing the regulation of exchanger activity are still unclear. This is especially important in the aggressively metastatic, triple-negative basal breast cancer subtype. We used affinity chromatography followed by mass spectrometry to identify novel and putative interaction partners of NHE1 in MDA-MB-231 triple-negative breast cancer cells. NHE1 associated with several types of proteins including cytoskeletal proteins and chaperones. We validated protein interactions by co-immunoprecipitation for: 14-3-3, AKT, α-enolase, CHP1, HSP70 and HSP90. Additionally, we used The Cancer Genome Atlas (TCGA) to study NHE1 gene expression in primary patient breast tumours versus adjacent normal tissue. NHE1 expression was elevated in breast tumour samples and, when broken down by breast cancer subtype, NHE1 gene expression was significantly lower in tumours of the basal subtype compared to luminal and HER2+ subtypes. Reverse phase protein array (RPPA) analysis showed that NHE1 expression positively correlated with p90RSK expression in basal, but not luminal, primary tumours. Other proteins were negatively correlated with NHE1 expression in basal breast cancer tumours. Taken together, our data provides the first insight into the signalling molecules that form the NHE1 interactome in triple-negative breast cancer cells. These results will focus our search for novel targeted therapies.


Subject(s)
Protein Interaction Mapping , Sodium-Hydrogen Exchanger 1/metabolism , Triple Negative Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Mass Spectrometry , Peptides/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Sodium-Hydrogen Exchanger 1/genetics , Triple Negative Breast Neoplasms/genetics
3.
Biochem Pharmacol ; 118: 31-39, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27521504

ABSTRACT

Hyper-activation of the Na+/H+ exchanger NHE1 occurs at the onset of oncogenic transformation and plays a critical role in breast cancer carcinogenesis. Dysregulation of NHE1 activity results in intracellular alkalinization and the acidification of the extracellular tumor microenvironment that promotes metastasis. Hence, the use of chemical inhibitors of NHE1 as chemotherapeutic agents is an alluring prospect. We previously demonstrated that two structurally different NHE1 inhibitors, EMD87580 [(2-methyl-4,5-di-(methylsulfonyl)-benzoyl)-guanidine], and HMA [5-(N,N-hexamethylene)-amiloride], were effective as co-adjuvants to potentiate paclitaxel-mediated cytotoxic chemotherapy in triple-negative breast cancer (TNBC) cells. Both these drugs, however, had reduced or minimal anti-cancer effects when used alone. Here, we tested KR-33028 (4-cyano (benzo[b]thiophene-2-carbonyl)guanidine), a potent and selective inhibitor of NHE1, to determine its efficacy in inhibition of metastatic potential of TNBC cells. In highly invasive MDA-MB-231, moderately invasive MDA-MB-468, and lowly invasive Hs578T TNBC cells, KR-33028 considerably reduced rates of cell migration and anchorage-independent colony growth. Invasion of MDA-MB-231 and MDA-MB-468 cells through extracellular matrix was also dramatically decreased in response to KR-33028. We further tested the effect of KR-33028 on MDA-MB-231 cells lacking NHE1 expression (231koNHE1); no differences were observed between untreated control and KR-33028-treated 231koNHE1 cells. Taken together, our results highlight the in vitro efficacy of KR-33028-mediated NHE1 inhibition on limiting cellular functions that are predictive of metastasis in vivo. We suggest that targeting NHE1 in the development of novel chemotherapeutics could be highly effective in combatting triple-negative breast cancer and that KR-33028 is potentially useful in prevention of metastasis.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Guanidines/pharmacology , Membrane Transport Modulators/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Thiophenes/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Intracellular Fluid/chemistry , Intracellular Fluid/drug effects , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Stem Cell Assay
5.
Oncotarget ; 7(16): 21091-113, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27049728

ABSTRACT

In triple-negative breast cancer (TNBC), the high recurrence rate, increased invasion and aggressive metastatic formation dictate patient survival. We previously demonstrated a critical role for the Na+/H+ exchanger isoform 1 (NHE1) in controlling metastasis of triple-negative cells. Here, we investigated the effect of changes to three regulatory loci of NHE1. Two via the Ras/Raf/ERK/p90RSK pathway: p90RSK/14-3-3 (S703A) and ERK1/2 (S766,770,771A, SSSA) and a third via a calmodulin-binding domain (K641,R643,645,647E, 1K3R4E). MDA-MB-231 cells with a mutation at the p90RSK site (S703A-NHE1) changed from a wild-type mesenchymal morphology to a smaller epithelial-like phenotype with a loss of expression of mesenchymal marker vimentin. S703A cells also had reduced metastatic potential and markedly decreased rates of migration, invasion, spheroid growth, anchorage-dependent and soft agar colony formation. Similarly, BI-D1870, a specific inhibitor of p90RSK, significantly inhibited the metastatic potential of highly invasive MDA-MB-231 and moderately invasive MDA-MB-468 TNBC cells, but was minimally effective in non-invasive Hs578T TNBC cells. In contrast, invasion and spheroid growth were unaffected in cells containing NHE1 with mutations interfering with its activation by ERK1/2 (SSSA), though rates of migration and colony formation were reduced. Cells with a constitutive activation of NHE1 via the 1K3R4E mutation exhibited higher rates of migration, invasion, and spheroid growth. Taken together, our data demonstrate the critical role of NHE1 in metastasis, and suggest a novel link between NHE1 and the expression and cytosolic organization of vimentin, a key factor in epithelial-mesenchymal transition, that is dependent on p90RSK/14-3-3-mediated activation of the exchanger.


Subject(s)
Biomarkers, Tumor/metabolism , Epithelial-Mesenchymal Transition , Sodium-Hydrogen Exchanger 1/metabolism , Triple Negative Breast Neoplasms/secondary , Apoptosis , Cell Adhesion , Cell Cycle , Cell Movement , Cell Proliferation , Female , Humans , Lymphatic Metastasis , Mutation , Sodium-Hydrogen Exchanger 1/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured
6.
Biochim Open ; 2: 16-23, 2016 Jun.
Article in English | MEDLINE | ID: mdl-29632834

ABSTRACT

Metastasis is the leading cause of mortality in patients with breast cancer. In triple-negative breast cancer, high recurrence rates, increased invasive capacity of cells, and their aggressive ability to metastasize at secondary sites dictate patient survival. The Na+/H+ exchanger isoform 1 (NHE1) plays a critical role in controlling the metastatic potential of these cells. Its activity results in an elevation of intracellular pH and in extracellular acidification, a key step in the establishment of the tumor microenvironment. Here, we describe assays for characterization of Na+/H+ exchanger activity and its related downstream physiological effects on triple-negative breast cancer cells. Na+/H+ exchanger activity can be routinely and rapidly measured in live cells with a fluorometric assay that assesses changes in intracellular pH. Characterization of downstream cell effector function as a result of Na+/H+ exchanger activation can be evaluated by measuring directed cell migration and invasion. Cell migration is assessed with wound-healing assays, where a gap is introduced in a confluent monolayer of cells and the rate of gap closure is measured over time. Cell invasion is assessed in the short-term by transwell invasion assays that track cell movement through an extracellular matrix. Long-term invasiveness, growth and proliferation can be assessed with 3-D invasion assays using transwell inserts fitted with specialized scaffolds optimized for 3-D cell culture. Taken together these assays provide powerful tools for testing the effects of altering Na+/H+ exchanger activity with chemical inhibition on the metastatic capacity of breast cancer cells.

7.
Oncotarget ; 6(2): 1262-75, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25514463

ABSTRACT

Dysregulation of Na⁺/H⁺ exchanger isoform one (NHE1) activity is a hallmark of cells undergoing tumorigenesis and metastasis, the leading cause of patient mortality. The acidic tumor microenvironment is thought to facilitate the development of resistance to chemotherapy drugs and to promote extracellular matrix remodeling leading to metastasis. Here, we investigated NHE1 as a co-adjuvant target in paclitaxel chemotherapy of metastatic breast cancer. We generated a stable NHE1-knockout of the highly invasive, triple-negative, MDA-MB-231 breast cancer cells. The NHE1-knockout cells proliferated comparably to parental cells, but had markedly lower rates of migration and invasion in vitro. In vivo xenograft tumor growth in athymic nude mice was also dramatically decreased compared to parental MDA-MB-231 cells. Loss of NHE1 expression also increased the susceptibility of knockout cells to paclitaxel-mediated cell death. NHE1 inhibition, in combination with paclitaxel, resulted in a dramatic decrease in viability, and migratory and invasive potential of triple-negative breast cancer cells, but not in hormone receptor-positive, luminal MCF7 cells. Our data suggest that NHE1 is critical in triple-negative breast cancer metastasis, and its chemical inhibition boosts the efficacy of paclitaxel in vitro, highlighting NHE1 as a novel, potential co-adjuvant target in breast cancer chemotherapy.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Paclitaxel/pharmacology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cation Transport Proteins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Female , Gene Knockout Techniques , Humans , MCF-7 Cells , Mice, Nude , Molecular Targeted Therapy/methods , Neoplasm Metastasis , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/genetics , Time Factors , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
8.
Cell Signal ; 26(6): 1355-68, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24583283

ABSTRACT

Insulin-induced insulin receptor (IR) tyrosine kinase activation and insulin cell survival responses have been reported to be under the regulation of a membrane associated mammalian neuraminidase-1 (Neu1). The molecular mechanism(s) behind this process is unknown. Here, we uncover a novel Neu1 and matrix metalloproteinase-9 (MMP-9) cross-talk in alliance with neuromedin B G-protein coupled receptor (GPCR), which is essential for insulin-induced IR activation and cellular signaling. Neu1, MMP-9 and neuromedin B GPCR form a complex with IRß subunit on the cell surface. Oseltamivir phosphate (Tamiflu®), anti-Neu1 antibodies, broad range MMP inhibitors piperazine and galardin (GM6001), MMP-9 specific inhibitor (MMP-9i), and GPCR neuromedin B specific antagonist BIM-23127 dose-dependently inhibited Neu1 activity associated with insulin stimulated rat hepatoma cells (HTCs) that overly express human IRs (HTC-IR). Tamiflu, anti-Neu1 antibodies and MMP-9i attenuated phosphorylation of IRß and insulin receptor substrate-1 (IRS1) associated with insulin-stimulated cells. Olanzapine, an antipsychotic agent associated with insulin resistance, induced Neu3 sialidase activity in WG544 or 1140F01 human sialidosis fibroblast cells genetically defective in Neu1. Neu3 antagonist 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (DANA) and anti-Neu3 antibodies inhibited sialidase activity associated with olanzapine treated murine Neu4 knockout macrophage cells. Olanzapine attenuated phosphorylation of IGF-R and IRS1 associated with insulin-stimulated human wild-type fibroblast cells. Our findings identify a novel insulin receptor-signaling platform that is critically essential for insulin-induced IRß tyrosine kinase activation and cellular signaling. Olanzapine-induced Neu3 sialidase activity attenuated insulin-induced IGF-R and IRS1 phosphorylation contributing to insulin resistance.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Neuraminidase/metabolism , Receptor, Insulin/metabolism , Signal Transduction , Animals , Antipsychotic Agents/pharmacology , Benzodiazepines/pharmacology , Cell Line, Tumor , Humans , Insulin/physiology , Insulin Receptor Substrate Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Olanzapine , Phosphorylation , Protein Processing, Post-Translational , Protein Transport , Rats , Receptor, IGF Type 1/metabolism
9.
Cell Signal ; 24(11): 2035-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22759791

ABSTRACT

The mechanism(s) behind GPCR transactivation of TLR receptors independent of TLR ligands is unknown. Here, GPCR agonists bombesin, bradykinin, lysophosphatidic acid (LPA), cholesterol, angiotensin-1 and -2, but not thrombin induce Neu1 activity in live macrophage cell lines and primary bone marrow macrophage cells from wild-type (WT) mice but not from Neu1-deficient mice. Using immunocytochemistry and NFκB-dependent secretory alkaline phosphatase (SEAP) analyses, bombesin induced NFκB activation in BMC-2 and RAW-blue macrophage cells, which was inhibited by MyD88 homodimerization inhibitor, Tamiflu, galardin, piperazine and anti-MMP-9 antibody. Bombesin receptor, neuromedin B (NMBR), forms a complex with TLR4 and MMP9. Silencing MMP9 mRNA using siRNA transfection of RAW-blue macrophage cells markedly reduced Neu1 activity associated with bombesin-, bradykinin- and LPA-treated cells to the untreated controls. These findings uncover a molecular organizational GPCR signaling platform to potentiate Neu1 and MMP-9 cross-talk on the cell surface that is essential for the transactivation of TLR receptors and subsequent cellular signaling.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Neuraminidase/metabolism , Receptors, G-Protein-Coupled/agonists , Toll-Like Receptors/metabolism , Animals , Antiviral Agents/pharmacology , Bombesin/pharmacology , Bradykinin/pharmacology , Cells, Cultured , Lysophospholipids/pharmacology , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Mice , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Neurokinin B/analogs & derivatives , Neurokinin B/metabolism , Oseltamivir/pharmacology , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Bombesin/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects , Vasodilator Agents/pharmacology
10.
J Biol Chem ; 286(42): 36532-49, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21873432

ABSTRACT

The signaling pathways of mammalian Toll-like receptors (TLRs) are well characterized, but the precise mechanism(s) by which TLRs are activated upon ligand binding remains poorly defined. Recently, we reported a novel membrane sialidase-controlling mechanism that depends on ligand binding to its TLR to induce mammalian neuraminidase-1 (Neu1) activity, to influence receptor desialylation, and subsequently to induce TLR receptor activation and the production of nitric oxide and proinflammatory cytokines in dendritic and macrophage cells. The α-2,3-sialyl residue of TLR was identified as the specific target for hydrolysis by Neu1. Here, we report a membrane signaling paradigm initiated by endotoxin lipopolysaccharide (LPS) binding to TLR4 to potentiate G protein-coupled receptor (GPCR) signaling via membrane Gα(i) subunit proteins and matrix metalloproteinase-9 (MMP9) activation to induce Neu1. Central to this process is that a Neu1-MMP9 complex is bound to TLR4 on the cell surface of naive macrophage cells. Specific inhibition of MMP9 and GPCR Gα(i)-signaling proteins blocks LPS-induced Neu1 activity and NFκB activation. Silencing MMP9 mRNA using lentivirus MMP9 shRNA transduction or siRNA transfection of macrophage cells and MMP9 knock-out primary macrophage cells significantly reduced Neu1 activity and NFκB activation associated with LPS-treated cells. These findings uncover a molecular organizational signaling platform of a novel Neu1 and MMP9 cross-talk in alliance with TLR4 on the cell surface that is essential for ligand activation of TLRs and subsequent cellular signaling.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Neuraminidase/metabolism , Signal Transduction/physiology , Toll-Like Receptor 4/metabolism , Animals , Cell Line , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Neuraminidase/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics
11.
Glycoconj J ; 27(6): 583-600, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20697956

ABSTRACT

Thymoquinone (TQ) derived from the nutraceutical black cumin oil has been reported to be a novel agonist of Neu4 sialidase activity in live cells (Glycoconj J DOI 10.1007/s10719-010-9281-6). The activation of Neu4 sialidase on the cell surface by TQ was found to involve GPCR-signaling via membrane targeting of Gαi subunit proteins and matrix metalloproteinase-9 activation. Contrary to other reports, TQ had no anti-inflammatory effects in vitro. Here, we show that MyD88/TLR4 complex formation and subsequent NFκB activation are induced by the Neu4 activity associated with TQ-stimulated live primary bone marrow (BM) macrophage cells from WT and Neu1-deficient mice, HEK-TLR4/MD2 cells and BMC-2 macrophage cell line but not with primary macrophage cells from Neu4-knockout mice. Tamiflu (oseltamivir phosphate), pertussis toxin (PTX), a specific inhibitor of Gαi proteins of G-protein coupled receptor (GPCR) and the broad range inhibitor of matrix metalloproteinase (MMP) galardin applied to live primary BM macrophage cells completely block TQ-induced MyD88/TLR4 complex formation. Using immunocytochemistry and western blot analyses, Tamiflu, galardin and PTX inhibit NFκB activation induced by Neu4 activity associated with TQ-stimulated BMC-2 cells, HEK-TLR4/MD2 cells and primary BM macrophages from WT mice. EMSA analyses on HEK-TLR4/MD2 nuclear cell extracts confirm the nuclear localization and DNA binding of TQ-induced NFκB activation in a biphasic manner within 30 min. Co-immunoprecipitation experiments reveal for the first time that MMP-9 may be an important intermediate link in the TQ-induced Neu4 activity circuitously targeting TLR4 receptors. Central to this process is that Neu4 forms a complex with MMP-9, which is already bound to TLR4 receptors. Fluorescence spectrophotometer analyses of live CD14-THP1 cells treated with TQ show Neu4 sialidase activity over 5 min. Using flow cytometry analyses, CD14-THP1 cells treated with TQ express stable protein levels of Neu4, TLR4 and MMP9 on the cell surface over 30 min except for a marked diminution of MMP9 at 15 min. Using cytokine array profiling analyses of serum, Neu4-knockout mice respond poorly to TQ in producing pro-inflammatory cytokines and chemokines after 5-h treatment compared to the wild-type or hypomorphic cathepsin A mice with a secondary 90% Neu1 deficient mice. Our findings establish an unprecedented signaling paradigm for TQ-induced Neu4 sialidase activity. It signifies that MMP-9 forms an important molecular signaling platform in complex with TLR4 receptors at the ectodomain and acts as the intermediate link for TQ-induced Neu4 sialidase in generating a functional receptor with subsequent NFκB activation and pro-inflammatory cytokine production in vivo.


Subject(s)
Benzoquinones/pharmacology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/enzymology , NF-kappa B/metabolism , Neuraminidase/biosynthesis , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Cytokines/blood , Dipeptides/pharmacology , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Humans , Lymphocyte Antigen 96/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Neuraminidase/antagonists & inhibitors , Oseltamivir/pharmacology , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Transport/drug effects , Receptors, G-Protein-Coupled/metabolism , Time Factors , Toll-Like Receptor 4/metabolism
12.
Glycoconj J ; 27(3): 329-48, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20213245

ABSTRACT

Anti-inflammatory activities of thymoquinone (TQ) have been demonstrated in in vitro and in vivo studies. However, the precise mechanism(s) of TQ in these anti-inflammatory activities is not well understood. Using a newly developed assay to detect sialidase activity in live macrophage cells (Glycoconj J doi: 10.1007/s10719-009-9239-8 ), here we show that TQ has no inhibitory effect on endotoxin lipopolysaccharide (LPS) induced sialidase activity in live BMC-2 macrophage cells. In contrast, the parent black seed oil (BSO) and another constituent of BSO para-cymene (p-CY) completely block LPS induced sialidase activity. All of these compounds had no effect on cell viability. On the other hand, TQ induces a vigorous sialidase activity in live BMC-2 macrophage cells in a dose dependent manner as well in live DC-2.4 dendritic cells, HEK-TLR4/MD2, HEK293, SP1 mammary adenocarcinoma cells, human WT and 1140F01 and WG0544 type I sialidosis fibroblast cells. Tamiflu (oseltamivir phosphate) inhibits TQ-induced sialidase activity in live BMC-2 cells with an IC(50) of 0.0194 microM compared to an IC(50) of 19.1 microM for neuraminidase inhibitor DANA (2-deoxy-2,3-dehydro-N-acetylneuraminic acid). Anti-Neu1, -2 and -3 antibodies have no inhibition of TQ-induced sialidase activity in live BMC-2 and human THP-1 macrophage cells but anti-Neu4 antibodies completely block this activity. There is a vigorous sialidase activity associated with TQ treated live primary bone marrow (BM) macrophage cells derived from WT and hypomorphic cathepsin A mice with a secondary Neu1 deficiency (NeuI KD), but not from Neu4 knockout (Neu4 KO) mice. Pertussis toxin (PTX), a specific inhibitor of Galphai proteins of G-protein coupled receptor (GPCR) and the broad range inhibitors of matrix metalloproteinase (MMP) galardin and piperazine applied to live BMC-2, THP-1 and primary BM macrophage cells completely block TQ-induced sialidase activity. These same inhibitory effects are not observed with the GM1 ganglioside specific cholera toxin subunit B (CTXB) as well as with CTX, tyrosine kinase inhibitor K252a, and the broad range GPCR inhibitor suramin. The specific inhibitor of MMP-9, anti-MMP-9 antibody and anti-Neu4 antibody, but not the specific inhibitor of MMP-3 completely block TQ-induced sialidase activity in live THP-1 cells, which express Neu4 and MMP-9 on the cell surface. Neu4 sialidase activity in cell lysates from TQ-treated live THP-1 cells desialylates natural gangliosides and mucin substrates. RT-PCR and western blot analyses reveal no correlation between mRNA and protein values for Neu3 and Neu4 in human monocytic THP-1 cells, suggesting for the first time a varied post-transcriptional mechanism for these two mammalian sialidases independent of TQ activation. Our findings establish an unprecedented activation of Neu4 sialidase on the cell surface by thymoquinone, which is derived from the nutraceutical black cumin oil. The potentiation of GPCR-signaling by TQ via membrane targeting of Galphai subunit proteins and matrix metalloproteinase-9 activation may be involved in the activation process of Neu4 sialidase on the cell surface.


Subject(s)
Benzoquinones/pharmacology , Fibroblasts/enzymology , Macrophages/enzymology , Matrix Metalloproteinase 9/metabolism , Mucolipidoses/enzymology , Neuraminidase/metabolism , Nigella sativa/chemistry , Receptors, G-Protein-Coupled/metabolism , Animals , Benzoquinones/chemistry , Blotting, Western , Cell Line , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Flow Cytometry , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Knockout , Polymerase Chain Reaction
13.
Cell Signal ; 22(8): 1193-205, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20347965

ABSTRACT

Neurotrophin-induced Trk tyrosine kinase receptor activation and neuronal cell survival responses have been reported to be under the control of a membrane associated sialidase. Here, we identify an unprecedented membrane sialidase mechanism initiated by nerve growth factor (NGF) binding to TrkA to potentiate GPCR-signaling via membrane Galphai subunit proteins and matrix metalloproteinase-9 (MMP-9) activation to induce Neu1 sialidase activation in live primary neurons and TrkA- and TrkB-expressing cell lines. Central to this process is that Neu1/MMP-9 complex is bound to TrkA on the cell surface of naïve primary neurons and TrkA-expressing cells. Tamiflu completely blocks this sialidase activity in live TrkA-PC12 cells treated with NGF with an IC(50) of 3.876 microM with subsequent inhibition of Trk activation in primary neurons and neurite outgrowth in TrkA-PC12 cells. Our findings uncover a Neu1 and MMP-9 cross-talk on the cell surface that is critically essential for neurotrophin-induced Trk tyrosine kinase receptor activation and cellular signaling.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Nerve Growth Factor/pharmacology , Neuraminidase/metabolism , Neurons/metabolism , Receptor, trkA/metabolism , Signal Transduction , Animals , Cell Membrane/enzymology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Mice , Neuraminidase/analysis , Neurons/enzymology , PC12 Cells , Rats
14.
Cell Signal ; 22(2): 314-24, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19796680

ABSTRACT

The ectodomain of TOLL-like receptors (TLR) is highly glycosylated with several N-linked gylcosylation sites located in the inner concave surface. The precise role of these sugar N-glycans in TLR receptor activation is unknown. Recently, we have shown that Neu1 sialidase and not Neu2, -3 and -4 forms a complex with TLR-2, -3 and -4 receptors on the cell-surface membrane of naïve and activated macrophage cells (Glycoconj J DOI 10.1007/s10719-009-9239-8). Activation of Neu1 is induced by TLR ligands binding to their respective receptors. Here, we show that endotoxin lipopolysaccharide (LPS)-induced MyD88/TLR4 complex formation and subsequent NFkappaB activation is dependent on the removal of alpha-2,3-sialyl residue linked to beta-galactoside of TLR4 by the Neu1 activity associated with LPS-stimulated live primary macrophage cells, macrophage and dendritic cell lines but not with primary Neu1-deficient macrophage cells. Exogenous alpha-2,3 sialyl specific neuraminidase (Streptoccocus pneumoniae) and wild-type T. cruzi trans-sialidase (TS) but not the catalytically inactive mutant TSAsp98-Glu mediate TLR4 dimerization to facilitate MyD88/TLR4 complex formation and NFkappaB activation similar to those responses seen with LPS. These same TLR ligand-induced NFkappaB responses are not observed in TLR deficient HEK293 cells, but are re-established in HEK293 cells stably transfected with TLR4/MD2, and are significantly inhibited by alpha-2,3-sialyl specific Maackia amurensis (MAL-2) lectin, alpha-2,3-sialyl specific galectin-1 and neuraminidase inhibitor Tamiflu but not by alpha-2,6-sialyl specific Sambucus nigra lectin (SNA). Taken together, the findings suggest that Neu1 desialylation of alpha-2,3-sialyl residues of TLR receptors enables in removing a steric hinderance to receptor association for TLR activation and cellular signaling.


Subject(s)
Neuraminidase/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glycoproteins/pharmacology , Glycosylation , Humans , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/metabolism , Neuraminidase/pharmacology , Oseltamivir/pharmacology , Phosphorylation , Toll-Like Receptor 4/chemistry
15.
Glycoconj J ; 26(9): 1197-212, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19430901

ABSTRACT

The signaling pathways of mammalian Toll-like receptors (TLR) are well characterized, but the initial molecular mechanisms activated following ligand interactions with the receptors remain poorly defined. Here, we show a membrane controlling mechanism that is initiated by ligand binding to TLR-2, -3 and-4 to induce Neu1 sialidase activity within minutes in live primary bone marrow (BM) macrophage cells and macrophage and dendritic cell lines. Central to this process is that Neu1 and not Neu2,-3 and-4 forms a complex with TLR-2,-3 and-4 on the cell surface of naïve macrophage cells. Neuraminidase inhibitors BCX1827, 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (DANA), zanamivir and oseltamivir carboxylate have a limited significant inhibition of the LPS-induced sialidase activity in live BMC-2 macrophage cells but Tamiflu (oseltamivir phosphate) completely blocks this activity. Tamiflu inhibits LPS-induced sialidase activity in live BMC-2 cells with an IC(50) of 1.2 microM compared to an IC(50) of 1015 microM for its hydrolytic metabolite oseltamivir carboxylate. Tamiflu blockage of LPS-induced Neu1 sialidase activity is not affected in BMC-2 cells pretreated with anticarboxylesterase agent clopidogrel. Endotoxin LPS binding to TLR4 induces Neu1 with subsequent activation of NFkappaB and the production of nitric oxide and pro-inflammatory IL-6 and TNFalpha cytokines in primary and macrophage cell lines. Hypomorphic cathepsin A mice with a secondary Neu1 deficiency respond poorly to LPS-induced pro-inflammatory cytokines compared to the wild-type or hypomorphic cathepsin A with normal Neu1 mice. Our findings establish an unprecedented mechanism for pathogen molecule-induced TLR activation and cell function, which is critically dependent on Neu1 sialidase activity associated with TLR ligand treated live primary macrophage cells and macrophage and dendritic cell lines.


Subject(s)
Dendritic Cells/enzymology , Macrophages/enzymology , Neuraminidase/metabolism , Receptors, Pattern Recognition/metabolism , Toll-Like Receptors/immunology , Animals , Bone Marrow Cells/cytology , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Survival/drug effects , Cells, Cultured , Cytokines/blood , Dendritic Cells/cytology , Dendritic Cells/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Inflammation Mediators/metabolism , Ligands , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Neuraminidase/antagonists & inhibitors , Neuraminidase/deficiency , Nitric Oxide/biosynthesis , Oseltamivir/pharmacology
16.
Glycobiology ; 17(7): 725-34, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17389653

ABSTRACT

Trypanosome trans-sialidase (TS) is a sialic acid-transferring enzyme and a novel ligand of tyrosine kinase (TrkA) receptors but not of neurotrophin receptor p75NTR. Here, we show that TS targets TrkB receptors on TrkB-expressing pheochromocytoma PC12 cells and colocalizes with TrkB receptor internalization and phosphorylation (pTrkB). Wild-type TS but not the catalytically inactive mutant TSDeltaAsp98-Glu induces pTrkB and mediates cell survival responses against death caused by oxidative stress in TrkA- and TrkB-expressing cells like those seen with nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF). These same effects are not observed in Trk deficient PC12(nnr5) cells, but are re-established in PC12(nnr5) cells stably transfected with TrkA or TrkB, are partially blocked by inhibitors of tyrosine kinase (K-252a), mitogen-activated protein/mitogen-activated kinase (PD98059) and completely blocked by LY294002, an inhibitor of phosphatidylinositol 3-kinase (PI3K). Both TrkA- and TrkB-expressing cells pretreated with TS or their natural ligands are protected against cell death caused by serum/glucose deprivation or from hypoxia-induced neurite retraction. The cell survival effects of NGF and BDNF against oxidative stress are significantly inhibited by the neuraminidase inhibitor, Tamiflu. Together, these observations suggest that trypanosome TS mimics neurotrophic factors in cell survival responses against oxidative stress, hypoxia-induced neurite retraction and serum/glucose deprivation.


Subject(s)
Glucose/metabolism , Glycoproteins/metabolism , Neuraminidase/metabolism , Oxidative Stress , Receptor, trkB/metabolism , Serum/metabolism , Trypanosoma cruzi/metabolism , Animals , Cell Survival , Enzyme Inhibitors/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hypoxia , Nerve Growth Factor/metabolism , Oseltamivir/pharmacology , PC12 Cells , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...