Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
FEBS Open Bio ; 14(7): 1192-1204, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38719785

ABSTRACT

Glioblastoma recruits various nontransformed cells from distant tissues. Although bone marrow-derived mesenchymal stem cells (MSCs) have been observed migrating to glioblastoma, the underlying mechanism driving MSC migration toward glioblastoma remains unclear. Tumor vascularity is critical in the context of recurrent glioblastoma and is closely linked to the expression of stromal cell-derived factor-1 (SDF-1). We demonstrated that cadherin-6 mediated MSC migration both toward SDF-1 and toward glioblastoma cells. Cadherin-6 knockdown resulted in the downregulation of MSCs capacity to migrate in response to SDF-1. Furthermore, MSCs with cadherin-6 knockdown exhibited impaired migration in response to conditioned media derived from glioblastoma cell lines (U87 and U373) expressing SDF-1, thus simulating the glioblastoma microenvironment. Moreover, MSCs enhanced the vasculogenic capacity of U87 cells without increasing the proliferation, cancer stem cell characteristics, or migration of U87. These results suggest that the current strategy of utilizing MSCs as carriers for antiglioblastoma drugs requires careful examination. Furthermore, cadherin-6 may represent a novel potential target for controlling the recruitment of MSCs toward glioblastoma.


Subject(s)
Cadherins , Cell Movement , Chemokine CXCL12 , Glioblastoma , Mesenchymal Stem Cells , Humans , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Mesenchymal Stem Cells/metabolism , Cadherins/metabolism , Cadherins/genetics , Cell Movement/genetics , Chemokine CXCL12/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Tumor Microenvironment
2.
Cell Death Discov ; 10(1): 19, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38212369

ABSTRACT

Mesenchymal stem cells are recruited from the bone marrow into breast tumors, contributing to the creation of a tumor microenvironment that fosters tropism for breast tumors. However, the intrinsic mechanisms underlying the recruitment of bone marrow-derived mesenchymal stem cells (MSCs) into the breast tumor microenvironment are still under investigation. Our discoveries identified zonula occludens-1 (ZO-1) as a specific intrinsic molecule that plays a vital role in mediating the collective migration of MSCs towards breast tumor cells and transforming growth factor beta (TGF-ß), which is a crucial factor secreted by breast tumor cells. Upon migration in response to MDA-MB-231 cells and TGF-ß, MSCs showed increased formation of adherens junction-like structures (AJs) expressing N-cadherin and α-catenin at their cell-cell contacts. ZO-1 was found to be recruited into the AJs at the cell-cell contacts between MSCs. Additionally, ZO-1 collaborated with α-catenin to regulate AJ formation, dependently on the SH3 and GUK domains of the ZO-1 protein. ZO-1 knockdown led to the impaired migration of MSCs in response to the stimuli and subsequent downregulation of AJs formation at the cell-cell contacts during MSCs migration. Overall, our study highlights the novel role of ZO-1 in guiding MSC migration towards breast tumor cells, suggesting its potential as a new strategy for controlling and re-engineering the breast tumor microenvironment.

3.
Mol Ther Nucleic Acids ; 31: 398-410, 2023 Mar 14.
Article in English | MEDLINE | ID: mdl-36817727

ABSTRACT

Alternative splicing of microexons (3-30 base pairs [bp]) is involved in important biological processes in brain development and human cancers. However, understanding a splicing process of non-3x bp microexons is scarce. We showed that 4 bp microexon of mitochondrial pyruvate carrier1 (MPC1) is constitutively included in mRNA. Based on our studies with minigene and exon island constructs, we found the strong exon definition region in the proximal introns bordering MPC1 microexon. Ultimately, we defined a nucleotide fragment from the 3'ss 67 bp of MPC1 microexon to the 5'ss consensus sequence, as a core exon island, which can concatenate its microexon and neighboring exons by splicing. Furthermore, we showed that insertion of the core exon island into a target exon or intron induced skip the target exon or enhance the splicing of an adjacent exon, respectively. Collectively, we suggest that the exon island derived from MPC1 microexon modifies genuine splicing patterns depending on its position, thereby providing insights on strategies for splicing-mediated gene correction.

4.
Br J Cancer ; 124(3): 634-644, 2021 02.
Article in English | MEDLINE | ID: mdl-33071283

ABSTRACT

BACKGROUND: Most cancer cells employ the Warburg effect to support anabolic growth and tumorigenesis. Here, we discovered a key link between Warburg effect and aberrantly activated Wnt/ß-catenin signalling, especially by pathologically significant APC loss, in CRC. METHODS: Proteomic analyses were performed to evaluate the global effects of KYA1797K, Wnt/ß-catenin signalling inhibitor, on cellular proteins in CRC. The effects of APC-loss or Wnt ligand on the identified enzymes, PKM2 and LDHA, as well as Warburg effects were investigated. A linkage between activation of Wnt/ß-catenin signalling and cancer metabolism was analysed in tumour of Apcmin/+ mice and CRC patients. The roles of PKM2 in cancer metabolism, which depends on Wnt/ß-catenin signalling, were assessed in xenograft-tumours. RESULTS: By proteomic analysis, PKM2 and LDHA were identified as key molecules regulated by Wnt/ß-catenin signalling. APC-loss caused the increased expression of metabolic genes including PKM2 and LDHA, and increased glucose consumption and lactate secretion. Pathological significance of this linkage was indicated by increased expression of glycolytic genes with Wnt target genes in tumour of Apcmin/+ mice and CRC patients. Warburg effect and growth of xenografted tumours-induced by APC-mutated-CRC cells were suppressed by PKM2-depletion. CONCLUSIONS: The ß-catenin-PKM2 regulatory axis induced by APC loss activates the Warburg effect in CRC.


Subject(s)
Carrier Proteins/metabolism , Colorectal Neoplasms/metabolism , Genes, APC , L-Lactate Dehydrogenase/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Thyroid Hormones/metabolism , Warburg Effect, Oncologic , Wnt Signaling Pathway , Animals , Carrier Proteins/genetics , Colorectal Neoplasms/genetics , Heterografts , Humans , L-Lactate Dehydrogenase/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasm Proteins/genetics , Proteomics , Thiazolidines/pharmacology , Thyroid Hormones/genetics , Tissue Array Analysis , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , Thyroid Hormone-Binding Proteins
5.
Biochem J ; 475(10): 1687-1699, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29669911

ABSTRACT

Mitochondrial pyruvate carrier (MPC), which is essential for mitochondrial pyruvate usage, mediates the transport of cytosolic pyruvate into mitochondria. Low MPC expression is associated with various cancers, and functionally associated with glycolytic metabolism and stemness. However, the mechanism by which MPC expression is regulated is largely unknown. In this study, we showed that MPC1 is down-regulated in human renal cell carcinoma (RCC) due to strong suppression of peroxisome proliferator-activated receptor-gamma co-activator (PGC)-1 alpha (PGC-1α). We also demonstrated that overexpression of PGC-1α stimulates MPC1 transcription, while depletion of PGC-1α by siRNA suppresses MPC expression. We found that PGC-1α interacts with estrogen-related receptor-alpha (ERR-α) and recruits it to the ERR-α response element motif located in the proximal MPC1 promoter, resulting in efficient activation of MPC1 expression. Furthermore, the MPC inhibitor, UK5099, blocked PGC-1α-induced pyruvate-dependent mitochondrial oxygen consumption. Taken together, our results suggest that MPC1 is a novel target gene of PGC-1α. In addition, low expression of PGC-1α in human RCC might contribute to the reduced expression of MPC, resulting in impaired mitochondrial respiratory capacity in RCC by limiting the transport of pyruvate into the mitochondrial matrix.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/genetics , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Energy Metabolism , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Monocarboxylic Acid Transporters , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Promoter Regions, Genetic , Pyruvic Acid/metabolism , Response Elements , Transcription Factors , Tumor Cells, Cultured
6.
Oncotarget ; 8(47): 82940-82955, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-29137314

ABSTRACT

The present study aimed to examine the associations between androgen receptor (AR) and forkhead box A1 (FOXA1) and to investigate clinicopathological features and survival according to both biomarker status in estrogen receptor (ER)-positive breast cancers using in vitro study, patient cohort data, and the cBioPortal for Cancer Genomics and Kaplan-Meier Plotter websites. Experiments using T47D and ZR75-1 demonstrated AR-overexpressing cell lines decreased in cell proliferation through downregulation of ER, but FOXA1 did not change. Knockdown of FOXA1 resulted in a significantly reduced cell viability. Patients with immunohistochemically AR(-)/FOXA1(-) tumor frequently showed node metastasis, high grade, and high Ki-67 proliferation, therefore, significantly worse survival in ER-positive disease. AR and FOXA1 mRNA levels were significantly higher in ER-positive than in ER-negative tumors and AR-low/FOXA1-low tumors showed high grade, frequent basal-like subtype and worse disease-free survival in ER-positive cancers of public gene dataset, similarly to patient cohort results. The Kaplan-Meier Plotter analysis independently validated patients with both low AR/FOXA1 tumor were significantly associated with worse relapse-free survival in ER-positive cancers. This study suggests that distinctive clinicopathological features according to AR and FOXA1 are determined and a lack of both biomarkers is an independent poor prognostic factor in ER-positive tumors.

7.
Biochem Biophys Res Commun ; 474(3): 547-553, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27114304

ABSTRACT

Clear cell renal carcinoma (RCC), the most common malignancy arising in the adult kidney, exhibits increased aerobic glycolysis and low mitochondrial respiration due to von Hippel-Lindau gene defects and constitutive hypoxia-inducible factor-α expression. Sirt3 is a major mitochondrial deacetylase that mediates various types of energy metabolism. However, the role of Sirt3 as a tumor suppressor or oncogene in cancer depends on cell types. We show increased Sirt3 expression in the mitochondrial fraction of human RCC tissues. Sirt3 depletion by lentiviral short-hairpin RNA, as well as the stable expression of the inactive mutant of Sirt3, inhibited cell proliferation and tumor growth in xenograft nude mice, respectively. Furthermore, mitochondrial pyruvate, which was used for oxidation in RCC, might be derived from glutamine, but not from glucose and cytosolic pyruvate, due to depletion of mitochondrial pyruvate carrier and the relatively high expression of malic enzyme 2. Depletion of Sirt3 suppressed glutamate dehydrogenase activity, leading to impaired mitochondrial oxygen consumption. Our findings suggest that Sirt3 plays a tumor-progressive role in human RCC by regulating glutamine-derived mitochondrial respiration, particularly in cells where mitochondrial usage of cytosolic pyruvate is severely compromised.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Proliferation , Glutamine/metabolism , Kidney Neoplasms/metabolism , Mitochondrial Proteins/metabolism , Sirtuin 3/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/pathology , Mitochondria/metabolism , Mitochondria/pathology , Oxidation-Reduction , Tumor Cells, Cultured
8.
FEBS Lett ; 588(17): 3074-80, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-24952355

ABSTRACT

Phosphoglucomutase (PGM)1 catalyzes the reversible conversion reaction between glucose-1-phosphate (G-1-P) and glucose-6-phosphate (G-6-P). Although both G-1-P and G-6-P are important intermediates for glucose and glycogen metabolism, the biological roles and regulatory mechanisms of PGM1 are largely unknown. In this study we found that T553 is obligatory for PGM1 stability and the last C-terminal residue, T562, is critical for its activity. Interestingly, depletion of PGM1 was associated with declined cellular glycogen content and decreased rates of glycogenolysis and glycogenesis. Furthermore, PGM1 depletion suppressed cell proliferation under long-term repetitive glucose depletion. Our results suggest that PGM1 is required for sustained cell growth during nutritional changes, probably through regulating the balance of G-1-P and G-6-P in order to satisfy the cellular demands during nutritional stress.


Subject(s)
Glucose/deficiency , Phosphoglucomutase/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Stability , Glucose/pharmacology , Glucose-6-Phosphate/metabolism , Glucosephosphates/metabolism , Glycogen/metabolism , Humans , Phosphoglucomutase/chemistry , Threonine
9.
Biochem J ; 453(1): 49-60, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23627357

ABSTRACT

PPARγ (peroxisome-proliferator-activated receptor γ) is a master transcription factor involved in adipogenesis through regulating adipocyte-specific gene expression. Recently, lipin1 was found to act as a key factor for adipocyte maturation and maintenance by modulating the C/EBPα (CCAAT/enhancer-binding protein α) and PPARγ network; however, the precise mechanism by which lipin1 affects the transcriptional activity of PPARγ is largely unknown. The results of the present study show that lipin1 activates PPARγ by releasing co-repressors, NCoR1 (nuclear receptor co-repressor 1) and SMRT (silencing mediator of retinoid and thyroid hormone receptor), from PPARγ in the absence of the ligand rosiglitazone. We also identified a novel lipin1 TAD (transcriptional activation domain), between residues 217 and 399, which is critical for the activation of PPARγ, but not PPARα. Furthermore, this TAD is unique to lipin1 since this region does not show any homology with the other lipin isoforms, lipin2 and lipin3. The activity of the lipin1 TAD is enhanced by p300 and SRC-1 (steroid receptor co-activator 1), but not by PCAF (p300/CBP-associated factor) and PGC-1α (PPAR co-activator 1α). The physical interaction between lipin1 and PPARγ occurs at the lipin1 C-terminal region from residues 825 to 926, and the VXXLL motif at residue 885 is critical for binding with and the activation of PPARγ. The action of lipin1 as a co-activator of PPARγ enhanced adipocyte differentiation; the TAD and VXXLL motif played critical roles, but the catalytic activity of lipin1 was not directly involved. Collectively, these data suggest that lipin1 functions as a key regulator of PPARγ activity through its ability to release co-repressors and recruit co-activators via a mechanism other than PPARα activation.


Subject(s)
Nuclear Proteins/physiology , PPAR gamma/genetics , Phosphatidate Phosphatase/physiology , 3T3-L1 Cells , Adipocytes/cytology , Animals , Cell Differentiation/drug effects , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , PPAR alpha/metabolism , PPAR gamma/metabolism , Transcription, Genetic/drug effects
10.
Proc Natl Acad Sci U S A ; 108(29): 11930-5, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730131

ABSTRACT

Contact inhibition of cell growth is essential for embryonic development and maintenance of tissue architecture in adult organisms, and the growth of tumors is characterized by a loss of contact inhibition of proliferation. The recently identified Hippo signaling pathway has been implicated in contact inhibition of proliferation as well as organ size control. The modulation of the phosphorylation and nuclear localization of Yes-associated protein (YAP) by the highly conserved kinase cascade of the Hippo signaling pathway has been intensively studied. However, cell-surface receptors regulating the Hippo signaling pathway in mammals are not well understood. In this study, we show that Hippo signaling pathway components are required for E-cadherin-dependent contact inhibition of proliferation. Knockdown of the Hippo signaling components or overexpression of YAP inhibits the decrease in cell proliferation caused by E-cadherin homophilic binding at the cell surface, independent of other cell-cell interactions. We also demonstrate that the E-cadherin/catenin complex functions as an upstream regulator of the Hippo signaling pathway in mammalian cells. Expression of E-cadherin in MDA-MB-231 cells restores the density-dependent regulation of YAP nuclear exclusion. Knockdown of ß-catenin in densely cultured MCF10A cells, which mainly depletes E-cadherin-bound ß-catenin, induces a decrease in the phosphorylation of S127 residue of YAP and its nuclear accumulation. Moreover, E-cadherin homophilic binding independent of other cell interactions is sufficient to control the subcellular localization of YAP. Therefore, Our results indicate that, in addition to its role in cell-cell adhesion, E-cadherin-mediated cell-cell contact directly regulates the Hippo signaling pathway to control cell proliferation.


Subject(s)
Cadherins/metabolism , Cell Proliferation , Contact Inhibition/physiology , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Cell Cycle Proteins , Cell Line , Drosophila , Drosophila Proteins/genetics , Fluorescent Antibody Technique, Indirect , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/genetics , Microspheres , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Staphylococcal Protein A/metabolism
11.
J Biol Chem ; 286(27): 23808-16, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21586797

ABSTRACT

Krüppel-like factor 4 (KLF4) is a transcription factor that plays an important role in cell differentiation, proliferation, and survival, especially in the context of cancers. This study revealed that KLF4 activates glycolytic metabolism in breast cancer cells by up-regulating the platelet isoform of phosphofructokinase (PFKP). KLF4 activated the transcription of the PFKP gene by directly binding to the PFKP promoter. Whereas glucose uptake and lactate production were inhibited by the knockdown of KLF4, they were activated by the overexpression of KLF4. Unlike PFKP, the expressions of the other isoforms of phosphofructokinase and glycolytic genes were unaffected by KLF4. The human breast cancer tissues showed a close correlation between KLF4 and PFKP expression. This study also showed that PFKP plays a critical role in cell proliferation in breast cancer cells. In conclusion, it is suggested that KLF4 plays a role in maintenance of high glycolytic metabolism by transcriptional activation of the PFKP gene in breast cancer cells.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Kruppel-Like Transcription Factors/metabolism , Phosphofructokinase-1, Type C/biosynthesis , Promoter Regions, Genetic , Transcription, Genetic , Breast Neoplasms , Cell Line, Tumor , Female , Glucose/genetics , Glucose/metabolism , Glycolysis/genetics , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Lactic Acid/metabolism , Phosphofructokinase-1, Type C/genetics
12.
PLoS One ; 4(12): e8411, 2009 Dec 22.
Article in English | MEDLINE | ID: mdl-20027292

ABSTRACT

BACKGROUND: Paraxial protocadherin (PAPC) and fibronectin leucine-rich domain transmembrane protein-3 (FLRT3) are induced by TGFbeta signaling in Xenopus embryos and both regulate morphogenesis by inhibiting C-cadherin mediated cell adhesion. PRINCIPAL FINDINGS: We have investigated the functional and physical relationships between PAPC, FLRT3, and C-cadherin. Although neither PAPC nor FLRT3 are required for each other to regulate C-cadherin adhesion, they do interact functionally and physically, and they form a complex with cadherins. By itself PAPC reduces cell adhesion physiologically to induce cell sorting, while FLRT3 disrupts adhesion excessively to cause cell dissociation. However, when expressed together PAPC limits the cell dissociating and tissue disrupting activity of FLRT3 to make it effective in physiological cell sorting. PAPC counteracts FLRT3 function by inhibiting the recruitment of the GTPase RND1 to the FLRT3 cytoplasmic domain. CONCLUSIONS/SIGNIFICANCE: PAPC and FLRT3 form a functional complex with cadherins and PAPC functions as a molecular "governor" to maintain FLRT3 activity at the optimal level for physiological regulation of C-cadherin adhesion, cell sorting, and morphogenesis.


Subject(s)
Cadherins/metabolism , Embryo, Nonmammalian/cytology , Membrane Proteins/metabolism , Morphogenesis , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Embryo, Nonmammalian/metabolism , Flow Cytometry , Gene Knockdown Techniques , Humans , Immunoprecipitation , Membrane Proteins/antagonists & inhibitors , Protein Binding , Protein Transport , Protocadherins , Xenopus Proteins/antagonists & inhibitors , Xenopus laevis/metabolism , rho GTP-Binding Proteins/metabolism
13.
Lipids Health Dis ; 8: 4, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19222837

ABSTRACT

BACKGROUND: The secreted enzyme autotaxin (ATX) stimulates tumor cell migration, tumorigenesis, angiogenesis, and metastasis. ATX hydrolyzes nucleotides, but its hydrolysis of lysophospholipids to produce lysophosphatidic acid (LPA) accounts for its biological activities. ATX has been identified only as a constitutively active enzyme, and regulation of its activity is largely unexplored. In spite of its presence in plasma along with abundant putative substrate LPC, the product LPA is found in plasma at unexpectedly low concentrations. It is plausible that the LPA-producing activity of ATX is regulated by its expression and by access to substrate(s). For this reason studying the interaction of enzyme with substrate is paramount to understanding the regulation of LPA production. RESULTS: In this study we determine ATX hydrolytic activities toward several artificial and natural substrates. Two novel point mutations near the enzyme active site (H226Q and H434Q) confer attenuated activity toward all substrates tested. The Vmax for LPC compounds depends upon chain length and saturation; but this order does not differ among wild type and mutants. However the mutant forms show disproportionately low activity toward two artificial substrates, pNpTMP and FS-3. The mutant forms did not significantly stimulate migration responses at concentrations that produced a maximum response for WT-ATX, but this defect could be rescued by inclusion of exogenous LPC. CONCLUSION: H226Q-ATX and H434Q-ATX are the first point mutations of ATX/NPP2 demonstrated to differentially impair substrate hydrolysis, with hydrolysis of artificial substrates being disproportionately lower than that of LPC. This implies that H226 and H434 are important for substrate interaction. Assays that rely on hydrolyses of artificial substrates (FS-3 and pNpTMP), or that rely on hydrolysis of cell-derived substrate, might fail to detect certain mutated forms of ATX that are nonetheless capable of producing LPA in the presence of sufficient exogenous substrate. H420Q-ATX could not be differentiated from WT-ATX, indicating that histidine at position 420 is not required for any of the activities of ATX tested in this study.


Subject(s)
Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutant Proteins/metabolism , Phosphodiesterase I/genetics , Phosphodiesterase I/metabolism , Point Mutation/genetics , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Amino Acid Substitution/drug effects , Cell Movement/drug effects , Fatty Acids/metabolism , Humans , Hydrolysis/drug effects , Immunoblotting , Kinetics , Lysophospholipids/pharmacology , Mutant Proteins/genetics , Phosphoric Diester Hydrolases , Substrate Specificity/drug effects
14.
Cell Signal ; 19(6): 1328-38, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17307336

ABSTRACT

Lysophosphatidic acid (LPA) stimulates sphingosine-1-phosphate (S1P)-sensitive motility in NIH3T3 clone7 cells. S1P inhibits motility only when added to the bottom well of the Boyden chamber, suggesting that pseudopodia can respond to their microenvironment. In order to study and localize this effect, we utilized a Transwell insert system to isolate pseudopodia. LPA stimulates protrusion of pseudopodia that are enriched in RhoA compared to cell bodies. Removal of LPA results in slow retraction with loss of vinculin-rich adhesion complexes and prolonged activation of RhoA. However, RhoA, ROCK and mDia are not required for this process. In contrast, rapid retraction, induced by adding S1P to the bottom well, is associated with a quick spike of activated RhoA and coalescence of adhesion complexes that colocalize with the ends of stress fibers. S1P-induced retraction requires RhoA and ROCK but is only delayed by inhibition of mDia. These data indicate that pseudopodia sense and integrate signals initiated by localized bioactive lipids, affecting both cellular polarity and their own function in motility.


Subject(s)
Lysophospholipids/pharmacology , Pseudopodia/drug effects , Pseudopodia/enzymology , Sphingosine/analogs & derivatives , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Cell Polarity/drug effects , Enzyme Activation/drug effects , Formins , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microtubules/drug effects , Microtubules/metabolism , NIH 3T3 Cells , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Sphingosine/pharmacology , Vinculin/metabolism , rac GTP-Binding Proteins/metabolism , rho-Associated Kinases , rhoA GTP-Binding Protein/antagonists & inhibitors
15.
Cancer Lett ; 250(1): 53-62, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17189669

ABSTRACT

The histone deacetylase inhibitor, trichostatin A (TSA), and the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (Aza-dC), induced epigenetic regulation of sphingosine-1-phosphate (S1P) receptors in human melanoma cells, switching S1P from motility inhibitor to stimulator. Quantitative PCR revealed increased expression of S1P(1) and S1P(3), associated with S1P-induced chemotaxis, and decreased expression of S1P(2), associated with motility inhibition. Expression of lysophosphatidic acid (LPA) receptors was less affected. The TSA effect was reversible suggesting no mutational change, and Aza-dC treatment resulted in demethylation of a putative S1P(1) promoter. S1P receptors, therefore, appear to be susceptible to epigenetic regulation, accompanied by altered cellular functionality.


Subject(s)
Azacitidine/analogs & derivatives , Cell Movement/drug effects , Epigenesis, Genetic , Hydroxamic Acids/pharmacology , Lysophospholipids/pharmacology , Melanoma/genetics , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation , Decitabine , Gene Expression Regulation , Humans , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Lysophospholipid , Sphingosine/pharmacology
16.
J Biol Chem ; 281(32): 22786-93, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16782709

ABSTRACT

Autotaxin (ATX, nucleotide pyrophosphate/phosphodiesterase-2) is an autocrine motility factor initially characterized from A2058 melanoma cell-conditioned medium. ATX is known to contribute to cancer cell survival, growth, and invasion. Recently ATX was shown to be responsible for the lysophospholipase D activity that generates lysophosphatidic acid (LPA). Production of LPA is sufficient to explain the effects of ATX on tumor cells. Cyclic phosphatidic acid (cPA) is a naturally occurring analog of LPA in which the sn-2 hydroxy group forms a 5-membered ring with the sn-3 phosphate. Cellular responses to cPA generally oppose those of LPA despite activation of apparently overlapping receptor populations, suggesting that cPA also activates cellular targets distinct from LPA receptors. cPA has previously been shown to inhibit tumor cell invasion in vitro and cancer cell metastasis in vivo. However, the mechanism governing this effect remains unresolved. Here we show that 3-carba analogs of cPA lack significant agonist activity at LPA receptors yet are potent inhibitors of ATX activity, LPA production, and A2058 melanoma cell invasion in vitro and B16F10 melanoma cell metastasis in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Multienzyme Complexes/chemistry , Phosphatidic Acids/chemistry , Phosphodiesterase I/chemistry , Pyrophosphatases/chemistry , Cell Line, Tumor , Culture Media, Conditioned , Humans , Lipid Metabolism , Lysophospholipids/pharmacology , Melanoma/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphoric Diester Hydrolases , Recombinant Proteins/chemistry , Spectrometry, Fluorescence
17.
Lipids Health Dis ; 4: 5, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15737239

ABSTRACT

BACKGROUND: Autotaxin (ATX, NPP-2), originally purified as a potent tumor cell motility factor, is now known to be the long-sought plasma lysophospholipase D (LPLD). The integrity of the enzymatic active site, including three crucial histidine moieties, is required for motility stimulation, as well as LPLD and 5'nucleotide phosphodiesterase (PDE) activities. Except for relatively non-specific chelation agents, there are no known inhibitors of the ATX LPLD activity. RESULTS: We show that millimolar concentrations of L-histidine inhibit ATX-stimulated but not LPA-stimulated motility in two tumor cell lines, as well as inhibiting enzymatic activities. Inhibition is reversed by 20-fold lower concentrations of zinc salt. L-histidine has no significant effect on the Km of LPLD, but reduces the Vmax by greater than 50%, acting as a non-competitive inhibitor. Several histidine analogs also inhibit the LPLD activity of ATX; however, none has greater potency than L-histidine and all decrease cell viability or adhesion. CONCLUSION: L-histidine inhibition of LPLD is not a simple stoichiometric chelation of metal ions but is more likely a complex interaction with a variety of moieties, including the metal cation, at or near the active site. The inhibitory effect of L-histidine requires all three major functional groups of histidine: the alpha amino group, the alpha carboxyl group, and the metal-binding imidazole side chain. Because of LPA's involvement in pathological processes, regulation of its formation by ATX may give insight into possible novel therapeutic approaches.


Subject(s)
Cytokines/pharmacology , Histidine/pharmacology , Lysophospholipids/biosynthesis , Multienzyme Complexes/pharmacology , Neoplasms/metabolism , Phosphodiesterase I/pharmacology , Pyrophosphatases/pharmacology , Cations, Divalent/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Enzyme Activation/drug effects , Histidine/analogs & derivatives , Humans , Molecular Structure , Neoplasms/pathology , Phosphoric Diester Hydrolases/metabolism , Substrate Specificity , Zinc/chemistry , Zinc/pharmacology
18.
Cancer Res ; 63(17): 5446-53, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500380

ABSTRACT

Autotaxin (ATX) is an exoenzyme that potently induces tumor cell motility, and enhances experimental metastasis and angiogenesis. ATX was shown recently to be identical to serum lysophospholipase D activity, producing lysophosphatidic acid (LPA) from lyso-glycerophospholipids. LPA, itself a strong chemoattractant for tumor cells, may mediate the actions of ATX. We now extend the substrate specificity to sphingosylphosphorylcholine (SPC), which ATX hydrolyzes to sphingosine-1-phosphate (S1P). Under migration assay conditions, this novel reaction for the production of S1P has a substrate (SPC) K(m) = 0.23 +/- 0.07 mM. In our responder cell lines (NIH3T3 clone7 and A2058), S1P exerts maximal biological effects at concentrations of 10-100 nM and is mimicked in its biological effects by ATX plus SPC. These effects include inhibition of ATX- and LPA-stimulated motility, and elevation of activated Rho. In NIH3T3 clone7 cells stimulated with platelet-derived growth factor and treated with 10-25 nM S1P, motility is not inhibited and activation of Rho is unaffected, indicating that S1P possesses specificity in its effects. The exoenzyme ATX can potentially regulate diverse processes such as motility and angiogenesis via the S1P family of receptors. Because ATX hydrolyzes nucleotides, lyso-glycerophospholipids, and phosphosphingolipids into bioactive products, it possesses the ability, depending on the availability of substrates, to act as positive or negative regulator of receptor-mediated activity in the cellular microenvironment.


Subject(s)
Glucose-6-Phosphate Isomerase/pharmacology , Glycoproteins/pharmacology , Lysophospholipids , Multienzyme Complexes , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/metabolism , Receptors, G-Protein-Coupled , Sphingosine/analogs & derivatives , Sphingosine/biosynthesis , Sphingosine/metabolism , 3T3 Cells , Animals , COS Cells , Catalysis , Cell Movement/physiology , Chlorocebus aethiops , Hydrolysis/drug effects , Mice , Phosphodiesterase I , Phosphoric Diester Hydrolases , Pyrophosphatases , Receptors, Cell Surface/biosynthesis , Receptors, Lysophospholipid , rho GTP-Binding Proteins/metabolism
19.
Cancer Res ; 63(9): 2042-5, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12727817

ABSTRACT

The exo-enzyme autotaxin/NPP2 (ATX/NPP2) is a potent stimulator of cell migration, invasion, metastasis, and angiogenesis. Recently, ATX/NPP2 was found to possess lysophospholipase D (lyso-LPD) activity, generating the bioactive mediator lysophosphatidic acid from precursors. In the present study, we used site-directed mutagenesis to delineate the active domain of lysophospholipid catalytic activity and to examine potential overlap with the nucleotide phosphodiesterase domain. We found four amino acid residues obligatory for the phosphodiesterase, lyso-PLD, and migration-stimulating activities of ATX/NPP2, suggesting that 5'-nucleotide phosphodiesterase (PDE) and lyso-PLD share a common reaction mechanism and inviting design of enzymatic inhibitors as therapeutic agents for neoplastic disease.


Subject(s)
Glucose-6-Phosphate Isomerase/genetics , Glucose-6-Phosphate Isomerase/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Multienzyme Complexes , Phosphoric Diester Hydrolases/metabolism , Point Mutation , Animals , COS Cells , Cell Movement/genetics , Chlorocebus aethiops , Humans , Mutagenesis, Site-Directed , Phosphodiesterase I , Phosphoric Diester Hydrolases/genetics , Protein Structure, Tertiary , Pyrophosphatases , Receptors, Purinergic P1/physiology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...