Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
FASEB J ; 34(12): 15888-15906, 2020 12.
Article in English | MEDLINE | ID: mdl-33047359

ABSTRACT

A successful acute inflammatory response results in the elimination of infectious agents by neutrophils and monocytes, followed by resolution and repair through tissue-resident and recruited macrophages. Resolvins (D-series and E-series) are pro-resolving lipid mediators involved in resolution and tissue repair, whose intracellular signaling remains of interest. Here, we report that D-series resolvins (RvD1- RvD5) activate phospholipase D (PLD), a ubiquitously expressed membrane lipase enzyme activity in modulating phagocyte functions. The mechanism for PLD-mediated actions of Resolvin-D5 (RvD5) in polarizing macrophages (M1-like toward M2-like) was found to be two-pronged: (a) RvD5 inhibits post-transcriptional modifications, by miRs and 3'exonucleases that process PLD2 mRNA, thus increasing PLD2 expression and activity; and (b) RvD5 enhances PLD2-S6Kinase signaling required for membrane expansion and efferocytosis. In an in vivo model of second organ reflow injury, we found that RvD5 did not reduce lung neutrophil myeloperoxidase levels in PLD2-/- mice compared to WT and PLD1-/- mice, confirming a novel role of PLD2 as the isoform in RvD5-mediated resolution processes. These results demonstrate that RvD5-PLD2 are attractive targets for therapeutic interventions in vascular inflammation such as ischemia-reperfusion injury and cardiovascular diseases.


Subject(s)
Docosahexaenoic Acids/pharmacology , Inflammation/metabolism , Phagocytes/drug effects , Phospholipase D/metabolism , Animals , Cells, Cultured , Female , Humans , Inflammation/drug therapy , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Lung/drug effects , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/metabolism , Phagocytes/metabolism , Phagocytosis/drug effects , Protein Processing, Post-Translational/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Signal Transduction/drug effects
2.
Handb Exp Pharmacol ; 259: 89-113, 2020.
Article in English | MEDLINE | ID: mdl-31541319

ABSTRACT

Lipids are key building blocks of biological membranes and are involved in complex signaling processes such as metabolism, proliferation, migration, and apoptosis. Extracellular signaling by growth factors, stress, and nutrients is transmitted through receptors that activate lipid-modifying enzymes such as the phospholipases, sphingosine kinase, or phosphoinositide 3-kinase, which then modify phospholipids, sphingolipids, and phosphoinositides. One such important enzyme is phospholipase D (PLD), which cleaves phosphatidylcholine to yield phosphatidic acid and choline. PLD isoforms have dual role in cells. The first involves maintaining cell membrane integrity and cell signaling, including cell proliferation, migration, cytoskeletal alterations, and invasion through the PLD product PA, and the second involves protein-protein interactions with a variety of binding partners. Increased evidence of elevated PLD expression and activity linked to many pathological conditions, including cancer, neurological and inflammatory diseases, and infection, has motivated the development of dual- and isoform-specific PLD inhibitors. Many of these inhibitors are reported to be efficacious and safe in cells and mouse disease models, suggesting the potential for PLD inhibitors as therapeutics for cancer and other diseases. Current knowledge and ongoing research of PLD signaling networks will help to evolve inhibitors with increased efficacy and safety for clinical studies.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Phosphatidic Acids , Phospholipase D/antagonists & inhibitors , Animals , Humans , Mice , Signal Transduction
3.
Hepatology ; 69(4): 1632-1647, 2019 04.
Article in English | MEDLINE | ID: mdl-30411380

ABSTRACT

Intrahepatic accumulation of bile acids (BAs) causes hepatocellular injury. Upon liver damage, a potent protective response is mounted to restore the organ's function. Epidermal growth factor receptor (EGFR) signaling is essential for regeneration after most types of liver damage, including cholestatic injury. However, EGFR can be activated by a family of growth factors induced during liver injury and regeneration. We evaluated the role of the EGFR ligand, amphiregulin (AREG), during cholestatic liver injury and regulation of AREG expression by BAs. First, we demonstrated increased AREG levels in livers from patients with primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC). In two murine models of cholestatic liver injury, bile duct ligation (BDL) and alpha-naphthyl-isothiocyanate (ANIT) gavage, hepatic AREG expression was markedly up-regulated. Importantly, Areg-/- mice showed aggravated liver injury after BDL and ANIT administration compared to Areg+/+ mice. Recombinant AREG protected from ANIT and BDL-induced liver injury and reduced BA-triggered apoptosis in liver cells. Oral BA administration induced ileal and hepatic Areg expression, and, interestingly, cholestyramine feeding reduced postprandial Areg up-regulation in both tissues. Most interestingly, Areg-/- mice displayed high hepatic cholesterol 7 α-hydroxylase (CYP7A1) expression, reduced serum cholesterol, and high BA levels. Postprandial repression of Cyp7a1 was impaired in Areg-/- mice, and recombinant AREG down-regulated Cyp7a1 mRNA in hepatocytes. On the other hand, BAs promoted AREG gene expression and protein shedding in hepatocytes. This effect was mediated through the farnesoid X receptor (FXR), as demonstrated in Fxr-/- mice, and involved EGFR transactivation. Finally, we show that hepatic EGFR expression is indirectly induced by BA-FXR through activation of suppressor of cytokine signaling-3 (SOC3). Conclusion: AREG-EGFR signaling protects from cholestatic injury and participates in the physiological regulation of BA synthesis.


Subject(s)
Amphiregulin/metabolism , Bile Acids and Salts/biosynthesis , Cholestasis, Intrahepatic/metabolism , Cholesterol 7-alpha-Hydroxylase/metabolism , Animals , ErbB Receptors/metabolism , Humans , Mice, Inbred C57BL
4.
Oncogene ; 38(8): 1309-1323, 2019 02.
Article in English | MEDLINE | ID: mdl-30305726

ABSTRACT

Cancer cells are in continuous communication with the surrounding microenvironment and this communication can affect tumor evolution. In this work, we show that phospholipase D2 (PLD2) was overexpressed in colon tumors and is secreted by cancer cells, inducing senescence in neighboring fibroblasts. This occurs through its lipase domain. Senescence induced by its product, phosphatidic acid, leads to a senescence-associated secretory phenotype (SASP) able to increase the stem properties of cancer cells. This increase in stemness occurs by Wnt pathway activacion. This closes a feedback loop in which senescence acts as a crosspoint for the generation of CSCs mediated by phospholipid metabolism. We also demonstrate the connexion of both phenomena in mouse models in vivo showing that a high PLD2 expression increased stemness and tumorigenesis. Thus, the patients with colon cancer show high levels of PLD2 and SASP factor genes expression correlating with Wnt pathway activation. Therefore, we demonstrate that tumor cell-secreted PLD2 contributes to tumor development by modifying the microenvironment, making it a possible therapeutic target for cancer treatment. This mechanism may also explain the high levels of Wnt pathway activation in colon cancer.


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Phospholipase D/genetics , Tumor Microenvironment/genetics , Animals , Cell Proliferation/genetics , Cellular Senescence , Colonic Neoplasms/epidemiology , Colonic Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice , Neoplastic Stem Cells/pathology , Wnt Signaling Pathway/genetics
5.
Methods Mol Biol ; 1835: 297-314, 2018.
Article in English | MEDLINE | ID: mdl-30109659

ABSTRACT

Phospholipase D (PLD), is a protein that breaks down phospholipids, maintaining structural integrity and remodeling of cellular or intracellular membranes, as well as mediating protein trafficking and cytoskeletal dynamics during cell motility. One of the reaction products of PLD action is phosphatidic acid (PA). PA is a mitogen involved in a large variety of physiological cellular functions, such as cell growth, cell cycle progression, and cell motility. We have chosen as cell models the leukocyte polymorphonuclear neutrophil and the macrophage as examples of cell motility. We provide a three-part method for targeting PLD genetically and pharmacologically to study its role in cell migration. In the first part, we begin with genetically deficient mice PLD1-KO and PLD2-KO. We describe bone marrow neutrophil (BMN) isolation; BMN is labeled fluorescently and can be used for studying tissue-damaging neutrophilia in ischemia-reperfusion injury (IRI). In the second part, we begin also with PLD1-KO and PLD2-KO and prepare bone marrow-derived macrophages (BMDM), first from monocytes and then inducing macrophage differentiation in culture with continuous incubation of cytokines. We use BMDM to find experimentally if PLD woul play a role in cholesterol phagocytosis, which is the first step in atherosclerosis progression. In the third part, we study PLD function in BMN and BMDM with PLD enzyme pharmacological inhibitors instead of genetically deficient mice, to ascertain the particular contributions of isoforms PLD1 and PLD2 on leukocyte function. By using the three-step thorough approach, we could understand the molecular underpinning of PLD in the pathological conditions indicated above, IRI-neutrophilia and atherosclerosis.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Targeting , Leukocytes/drug effects , Leukocytes/physiology , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , Animals , Cell Movement/drug effects , Cell Movement/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Gene Targeting/methods , Isoenzymes , Macrophages/drug effects , Macrophages/physiology , Mice , Mice, Knockout , Multigene Family , Neutrophils/drug effects , Neutrophils/physiology , Phospholipase D/metabolism , RAW 264.7 Cells , Signal Transduction
6.
Cancer Metastasis Rev ; 37(2-3): 491-507, 2018 09.
Article in English | MEDLINE | ID: mdl-30091053

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is difficult to treat since cells lack the three receptors (ES, PR, or HER) that the most effective treatments target. We have used a well-established TNBC cell line (MDA-MB-231) from which we found evidence in support for a phospholipase D (PLD)-mediated tumor growth and metastasis: high levels of expression of PLD, as well as the absence of inhibitory miRs (such as miR-203) and 3'-mRNA PARN deadenylase activity in these cells. Such findings are not present in a luminal B cell line, MCF-7, and we propose a new miR•PARN•PLD node that is not uniform across breast cancer molecular subtypes and as such TNBC could be pharmacologically targeted differentially. We review the participation of PLD and phosphatidic acid (PA), its enzymatic product, as new "players" in breast cancer biology, with the aspects of regulation of the tumor microenvironment, macrophage polarization, regulation of PLD transcripts by specific miRs and deadenylases, and PLD-regulated exosome biogenesis. A new signaling miR•PARN•PLD node could serve as new biomarkers for TNBC abnormal signaling and metastatic disease staging, potentially before metastases are able to be visualized using conventional imaging.


Subject(s)
Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Phospholipase D/genetics , Phospholipase D/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Biomarkers, Tumor , Disease Progression , Exoribonucleases/metabolism , Female , Humans , MicroRNAs/genetics , Protein Biosynthesis , RNA, Untranslated/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
7.
J Leukoc Biol ; 103(5): 867-883, 2018 05.
Article in English | MEDLINE | ID: mdl-29656494

ABSTRACT

The uptake of cholesterol carried by low-density lipoprotein (LDL) is tightly controlled in the body. Macrophages are not well suited to counteract the cellular consequences of excess cholesterol leading to their transformation into "foam cells," an early step in vascular plaque formation. We have uncovered and characterized a novel mechanism involving phospholipase D (PLD) in foam cell formation. Utilizing bone marrow-derived macrophages from genetically PLD deficient mice, we demonstrate that PLD2 (but not PLD1)-null macrophages cannot fully phagocytose aggregated oxidized LDL (Agg-Ox-LDL), which was phenocopied with a PLD2-selective inhibitor. We also report a role for PLD2 in coupling Agg-oxLDL phagocytosis with WASP, Grb2, and Actin. Further, the clearance of LDL particles is mediated by both CD36 and PLD2, via mutual dependence on each other. In the absence of PLD2, CD36 does not engage in Agg-Ox-LDL removal and when CD36 is blocked, PLD2 cannot form protein-protein heterocomplexes with WASP or Actin. These results translated into humans using a GEO database of microarray expression data from atheroma plaques versus normal adjacent carotid tissue and observed higher values for NFkB, PLD2 (but not PLD1), WASP, and Grb2 in the atheroma plaques. Human atherectomy specimens confirmed high presence of PLD2 (mRNA and protein) as well as phospho-WASP in diseased arteries. Thus, PLD2 interacts in macrophages with Actin, Grb2, and WASP during phagocytosis of Agg-Ox-LDL in the presence of CD36 during their transformation into "foam cells." Thus, this study provides new molecular targets to counteract vascular plaque formation and atherogenesis.


Subject(s)
CD36 Antigens/metabolism , Foam Cells/pathology , Lipoproteins, LDL/metabolism , Phagocytosis , Phospholipase D/physiology , Plaque, Atherosclerotic/pathology , Animals , CD36 Antigens/genetics , Cells, Cultured , Cholesterol/metabolism , Female , Foam Cells/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
8.
J Leukoc Biol ; 103(5): 919-932, 2018 05.
Article in English | MEDLINE | ID: mdl-29437245

ABSTRACT

Phospholipase D (PLD) plays important roles in cellular responses to tissue injury that are critical to acute inflammatory diseases, such as the acute respiratory distress syndrome (ARDS). We investigated the expression of PLD isoforms and related phospholipid phosphatases in patients with ARDS, and their roles in a murine model of self-limited acute lung injury (ALI). Gene expression microarray analysis on whole blood obtained from patients that met clinical criteria for ARDS and clinically matched controls (non-ARDS) demonstrated that PLD1 gene expression was increased in patients with ARDS relative to non-ARDS and correlated with survival. In contrast, PLD2 expression was associated with mortality. In a murine model of self-resolving ALI, lung Pld1 expression increased and Pld2 expression decreased 24 h after intrabronchial acid. Total lung PLD activity was increased 24 h after injury. Pld1-/- mice demonstrated impaired alveolar barrier function and increased tissue injury relative to WT and Pld2-/- , whereas Pld2-/- mice demonstrated increased recruitment of neutrophils and macrophages, and decreased tissue injury. Isoform-specific PLD inhibitors mirrored the results with isoform-specific Pld-KO mice. PLD1 gene expression knockdown in human leukocytes was associated with decreased phagocytosis by neutrophils, whereas reactive oxygen species production and phagocytosis decreased in M2-macrophages. PLD2 gene expression knockdown increased neutrophil and M2-macrophage transmigration, and increased M2-macrophage phagocytosis. These results uncovered selective regulation of PLD isoforms after ALI, and opposing effects of selective isoform knockdown on host responses and tissue injury. These findings support therapeutic strategies targeting specific PLD isoforms for the treatment of ARDS.


Subject(s)
Acute Lung Injury/immunology , Leukocytes/immunology , Lung/immunology , Phospholipase D/physiology , Respiratory Distress Syndrome/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Case-Control Studies , Cells, Cultured , Female , Humans , Leukocytes/metabolism , Leukocytes/pathology , Lung/metabolism , Lung/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Phagocytosis , Protein Isoforms , Respiratory Distress Syndrome/pathology
9.
Adv Biol Regul ; 68: 107-119, 2018 05.
Article in English | MEDLINE | ID: mdl-28964725

ABSTRACT

Phospholipase D (PLD) plays a key role in both cell membrane lipid reorganization and architecture, as well as a cell signaling protein via the product of its enzymatic reaction, phosphatidic acid (PA). PLD is involved in promoting breast cancer cell growth, proliferation, and metastasis and both gene and protein expression are upregulated in breast carcinoma human samples. In spite of all this, the ultimate reason as to why PLD expression is high in cancer cells vs. their normal counterparts remains largely unknown. Until we understand this and the associated signaling pathways, it will be difficult to establish PLD as a bona fide target to explore new potential cancer therapeutic approaches. Recently, our lab has identified several molecular mechanisms by which PLD expression is high in breast cancer cells and they all involve post-transcriptional control of its mRNA. First, PA, a mitogen, functions as a protein and mRNA stabilizer that counteracts natural decay and degradation. Second, there is a repertoire of microRNAs (miRs) that keep PLD mRNA translation at low levels in normal cells, but their effects change with starvation and during endothelial-to-mesenchymal transition (EMT) in cancer cells. Third, there is a novel way of post-transcriptional regulation of PLD involving 3'-exonucleases, specifically the deadenylase, Poly(A)-specific Ribonuclease (PARN), which tags mRNA for mRNA for degradation. This would enable PLD accumulation and ultimately breast cancer cell growth. We review in depth the emerging field of post-transcriptional regulation of PLD, which is only recently beginning to be understood. Since, surprisingly, so little is known about post-transcriptional regulation of PLD and related phospholipases (PLC or PLA), this new knowledge could help our understanding of how post-transcriptional deregulation of a lipid enzyme expression impacts tumor growth.


Subject(s)
MicroRNAs/metabolism , Phospholipase D/metabolism , RNA, Messenger/metabolism , Ribonucleases/metabolism , Animals , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , MicroRNAs/genetics , Phospholipase D/genetics , RNA, Messenger/genetics , Ribonucleases/genetics , Signal Transduction/genetics , Signal Transduction/physiology
10.
Biol Open ; 6(2): 176-186, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28011629

ABSTRACT

The removal of mRNA transcript poly(A) tails by 3'→5' exonucleases is the rate-limiting step in mRNA decay in eukaryotes. Known cellular deadenylases are the CCR4-NOT and PAN complexes, and poly(A)-specific ribonuclease (PARN). The physiological roles and regulation for PARN is beginning to be elucidated. Since phospholipase D (PLD2 isoform) gene expression is upregulated in breast cancer cells and PARN is downregulated, we examined whether a signaling connection existed between these two enzymes. Silencing PARN with siRNA led to an increase in PLD2 protein, whereas overexpression of PARN had the opposite effect. Overexpression of PLD2, however, led to an increase in PARN expression. Thus, PARN downregulates PLD2 whereas PLD2 upregulates PARN. Co-expression of both PARN and PLD2 mimicked this pattern in non-cancerous cells (COS-7 fibroblasts) but, surprisingly, not in breast cancer MCF-7 cells, where PARN switches from inhibition to activation of PLD2 gene and protein expression. Between 30 and 300 nM phosphatidic acid (PA), the product of PLD enzymatic reaction, added exogenously to culture cells had a stabilizing role of both PARN and PLD2 mRNA decay. Lastly, by immunofluorescence microscopy, we observed an intracellular co-localization of PA-loaded vesicles (0.1-1 nm) and PARN. In summary, we report for the first time the involvement of a phospholipase (PLD2) and PA in mediating PARN-induced eukaryotic mRNA decay and the crosstalk between the two enzymes that is deregulated in breast cancer cells.

11.
PLoS One ; 11(11): e0166553, 2016.
Article in English | MEDLINE | ID: mdl-27851813

ABSTRACT

Phospholipase D-2 (PLD2) has a key role in breast cancer formation and metastasis formation with PLD small inhibitors reducing primary tumor growth. This study aimed to evaluate the importance of targeting PLD on the tumor microenvironment. We provide evidence about the beneficial effect of PLD inhibitors [FIPI (dual PLD1/PLD2) or VU0155072-2 (PLD2 inhibitor)] on avoiding infiltration of tumor-helping macrophages and neutrophils. Tumor growth and metastasis within the primary tumors had low (<20% over controls) PLD enzyme activity. Unexpectedly, we found that the inhibitors also affected PLD2 gene expression and protein albeit at a lesser extent. The later could indicate that targeting both the actual PLD enzyme and its activity could be beneficial for potential cancer treatments in vivo. F4/80 and Ly6G staining of macrophages and neutrophils, respectively, and Arg1 staining data were consistent with M2 and N2 polarization. NOS2 staining increased in xenotransplants upon treatment with PLD2 inhibitors suggesting the novel observation that an increased recruitment of M1 macrophages occurred in primary tumors. PLD inhibitor-treated primary tumors had large, fragile, necrotic areas that were Arg1+ for M2 macrophages. The xenotransplants also caused the formation of large F4/80+ and Ly6G+ (>100 µm) clusters in lungs. However, PLD inhibitors, particularly FIPI, were able to diminish leukocyte presence. Ex vivo chemotaxis and PLD activity of peripheral blood neutrophils (PMN) and peritoneal macrophages was also determined. Whereas PMN had impaired functionality, macrophages did not. This significantly increased ("emboldened") macrophage function was due to PLD inhibition. Since tumor-associated leukocytes in primary tumors and metastases were targeted via PLD inhibition, we posit that these inhibitors have a key role in cancer regression, while still affording an appropriate inflammatory response at least from off-site innate immunity macrophages.


Subject(s)
Breast Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Macrophages/pathology , Neutrophils/pathology , Phospholipase D/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Antigens, Ly/metabolism , Arginase/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Polarity/drug effects , Cellular Microenvironment/drug effects , Chemotaxis/drug effects , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunity, Innate/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/pathology , Macrophages/drug effects , Macrophages/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Neutrophils/drug effects , Neutrophils/metabolism , Nitric Oxide Synthase Type II/metabolism , Phospholipase D/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Oncotarget ; 7(30): 47002-47017, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27256981

ABSTRACT

The intracellular concentration of the mitogen phosphatidic acid (PA) must be maintained at low levels until the need arises for cell proliferation. How temporal and spatial trafficking of PA affects its target proteins in the different cellular compartments is not fully understood. We report that in cancer cells, PA cycles back and forth from the cellular membrane to the nucleus, affecting the function of epidermal growth factor (EGF), in a process that involves PPARα/LXRα signaling. Upon binding to its ligand, EGF receptor (EGFR)-initiated activation of phospholipase D (PLD) causes a spike in intracellular PA production that forms vesicles transporting EGFR from early endosomes (EEA1 marker) and prolonged internalization in late endosomes and Golgi (RCAS marker). Cells incubated with fluorescent-labeled PA (NBD-PA) show PA in "diffuse" locations throughout the cytoplasm, punctae (small, <0.1 µm) vesicles) and large (>0.5 µm) vesicles that co-localize with EGFR. We also report that PPARα/LXRα form heterodimers that bind to new Responsive Elements (RE) in the EGFR promoter. Nuclear PA enhances EGFR expression, a role compatible with the mitogenic ability of the phospholipid. Newly made EGFR is packaged into PA recycling vesicles (Rab11 marker) and transported back to the cytoplasm and plasma membrane. However, a PLD+PA combination impedes binding of PPARα/LXRα to the EGFR promoter. Thus, if PA levels inside the nucleus reach a certain threshold (>100 nM) PA outcompetes the nuclear receptors and transcription is inhibited. This new signaling function of PLD-PA targeting EGFR trafficking and biphasically modulating its transcription, could explain cell proliferation initiation and its maintenance in cancer cells.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , ErbB Receptors/metabolism , Phosphatidic Acids/metabolism , Signal Transduction/physiology , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Endosomes/metabolism , Epidermal Growth Factor/metabolism , Humans , Liver X Receptors/metabolism , PPAR alpha/metabolism , Phospholipase D/metabolism , Promoter Regions, Genetic , Protein Transport/physiology , Rats
13.
Mol Cell Biol ; 36(7): 1078-89, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26787840

ABSTRACT

We report a negative feedback loop between the signaling protein phospholipase D (PLD), phosphatidic acid (PA), and a specific set of microRNAs (miRNAs) during nutrient starvation of breast cancer cells. We show that PLD expression is increased in four breast cancer cell lines and that hypoxia, cell overcrowding, and nutrient starvation for 3 to 6 h increase expression even further. However, after prolonged (>12-h) starvation, PLD levels return to basal or lower levels. The mechanism for this is as follows. First, during initial starvation, an elevated PA (the product of PLD enzymatic activity) activates mTOR and S6K, known to inhibit apoptosis, and enhances cell migration especially in post-epithelial-to-mesenchymal transition (post-EMT) cancer cells. Second, continued PA production in later starvation induces expression of PLD-targeting microRNA 203 (miR-203), miR-887, miR-3619-5p, and miR-182, which reduce PLD translation. We provide direct evidence for a feedback loop, whereby PLD induction upon starvation leads to PA, which induces expression of miRNAs, which in turn inhibits PLD2 translation. The physiological relevance for breast cancer cells is that as PA can activate cell invasion, then, due to the negative feedback, it can deprive mTOR and S6K of their natural activator. It can further prevent inhibition of apoptosis and allow cells to survive nutrient deprivation, which normal cells cannot do.


Subject(s)
Breast Neoplasms/metabolism , Feedback, Physiological , MicroRNAs/metabolism , Phospholipase D/metabolism , Apoptosis , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Phosphatidic Acids/metabolism , Phospholipase D/genetics , Protein Biosynthesis , RNA, Neoplasm/metabolism , Ribosomal Protein S6/metabolism , TOR Serine-Threonine Kinases/metabolism
14.
Mol Oncol ; 10(5): 663-76, 2016 05.
Article in English | MEDLINE | ID: mdl-26781944

ABSTRACT

Slug (SNAI2) and Snail (SNAI1) are master regulatory transcription factors for organogenesis and wound healing, and they are involved in the epithelial to mesenchymal transition (EMT) of cancer cells. We found that the activity of phospholipase D isoform 2 (PLD2) is highly increased in cancers with larger size and poor prognosis (MDA-MB-231 versus MCF-7 cells), so we determined if Snail or Slug were responsible for PLD2 gene transcription regulation. Unexpectedly, we found that PLD2 expression was positively regulated by Slug but negatively regulated by Snail. The differential effects are amplified in breast cancer cells over normal cells and with MDA-MB-231 more robustly than MCF-7. Slug putatively binds to the PLD2 promoter and transactivates it, which is negated when Slug and Snail compete with each other. Meanwhile, PLD2 has a negative effect on Snail expression and a positive effect on Slug, thus closing a feedback loop between the lipase and the transcription factors. Further, PA, the product of PLD2 enzymatic reaction, has profound effects on its own and it further regulates the transcription factors. Thus, we show for the first time that the overexpressed PLD2 in human breast tumors is regulated by Slug and Snail transcription factors. The newly uncovered feedback loops in highly invasive cancer cells have important implications in the process of EMT.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Phospholipase D/genetics , Snail Family Transcription Factors/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic , Transcriptional Activation
15.
J Biol Chem ; 291(2): 719-30, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26567912

ABSTRACT

Breast cancer is a leading cause of morbidity and mortality among women. Metastasis is initiated after epithelial-mesenchymal-transition (EMT). We have found a connection between EMT markers and the expression of four microRNAs (miRs) mediated by the signaling enzyme phospholipase D (PLD). Low aggressive MCF-7 breast cancer cells have low endogenous PLD enzymatic activity and cell invasion, concomitant with high expression of miR-203, -887, and -3619 (that decrease PLD2 translation and a luciferase reporter) and miR-182 (targeting PLD1) that are, therefore, "tumor-suppressor-like" miRs. The combination miR-887+miR-3619 abolished >90% of PLD enzymatic activity. Conversely, post-EMT MDA-MB-231 cells have low miR expression, high levels of PLD1/2, and high aggressiveness. The latter was reversed by ectopically transfecting the miRs, which was negated by silencing miRs with specific siRNAs. We determined that the molecular mechanism is that E-cadherin triggers expression of the miRs in pre-EMT cells, whereas vimentin dampens expression of the miRs in post-EMT invasive cells. This novel work identifies for the first time a set of miRs that are activated by a major pre-EMT marker and deactivated by a post-EMT marker, boosting the transition from low invasion to high invasion, as mediated by the key phospholipid metabolism enzyme PLD.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Down-Regulation/genetics , MicroRNAs/genetics , Phospholipase D/metabolism , Vimentin/metabolism , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Conserved Sequence/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , MicroRNAs/metabolism , Models, Biological , Molecular Sequence Data , Neoplasm Invasiveness , Phenotype , Phospholipase D/genetics , Plasmids/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Sci Rep ; 5: 15379, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26493292

ABSTRACT

The expression of the epidermal growth factor receptor (EGFR) is highly regulated in normal cells, whereas some cancer cells have high constitutive levels. Understanding naturally-occurring ways of downregulating EGFR in cancer cells was investigated. Phosphatidic acid (PA) or Nuclear Receptors (NR) PPARα/RXRα/LXRα, enhance EGFR expression, mediated by the promoter region -856(A) to -226(T). Unexpectedly, the combination of NRs and PA caused repression. PA induces a conformational change in the nuclear receptor PPARα (increase of alpha-helices at the expense of decreasing beta-sheets), as evidenced by circular dichroism. This represses the naturally-enhancing capability of PPARα on EGFR transcription. PPARα-overexpressing cells in the presence of PA > 300 nM or the enzyme that produces it, phospholipase D (PLD), downregulate EGFR expression. The reasons are two-fold. First, PA displaces PPARα binding to the EGFR promoter at those concentrations. Second, NR heterodimer-dependent promoter activity is weakened in the presence of PA in vivo. Since other genes considered (ß-catenin, cyclin D3, PLD2 and ACOX-1) are also downregulated with a PA + PPARα combination, the transrepression appears to be a global phenomenon. Lastly, the reported effect is greater in MCF-7 than in MDA-MB-231 breast cancer cells, which could provide a novel basis for regulating excessive expression of EGFR in luminal cancer cells.


Subject(s)
ErbB Receptors/genetics , Orphan Nuclear Receptors/metabolism , PPAR alpha/metabolism , Phosphatidic Acids/pharmacology , Promoter Regions, Genetic , Base Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Circular Dichroism , DNA , Female , Humans , Liver X Receptors , Molecular Sequence Data
17.
Mol Cell Biol ; 35(18): 3131-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26124282

ABSTRACT

Overexpression of epidermal growth factor receptor (EGFR) is one of the frequent mechanisms implicated in cancer progression, and so is the overexpression of the enzyme phospholipase D (PLD) and its reaction product, phosphatidic acid (PA). However, an understanding of how these signaling molecules interact at the level of gene expression is lacking. Catalytically active PLD enhanced expression of EGFR in human breast cancer cells. Overexpression of the PLD2 isoform increased EGFR mRNA and protein expression. It also negated an EGFR downregulation mediated by small interfering RNA targeting EGFR (siEGFR). Several mechanisms contributed to the alteration in EGFR expression. First was the stabilization of EGFR transcripts as PLD2 delayed mRNA decay, which prolonged their half-lives. Second, RNase enzymatic activity was inhibited by PA. Third, protein stabilization also occurred, as indicated by PLD resistance to cycloheximide-induced EGFR protein degradation. Fourth, PA inhibited lysosomal and proteasomal degradation of internalized EGFR. PLD2 and EGFR colocalized at the cell membrane, and JAK3 phosphorylation at Tyr980/Tyr981 followed receptor endocytosis. Further, the presence of PLD2 increased stabilization of intracellular EGFR in large recycling vesicles at ∼15 min of EGF stimulation. Thus, PLD2-mediated production of PA contributed to the control of EGFR exposure to ligand through a multipronged transcriptional and posttranscriptional program during the out-of-control accumulation of EGFR signaling in cancer cells.


Subject(s)
ErbB Receptors/metabolism , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , Proteolysis/drug effects , RNA Stability/drug effects , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cycloheximide/pharmacology , Endocytosis , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Female , Humans , Janus Kinase 3/metabolism , Lysosomes/metabolism , MCF-7 Cells , Phospholipase D/biosynthesis , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering , Ribonucleases/antagonists & inhibitors , Signal Transduction
18.
PLoS Pathog ; 11(3): e1004696, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25768646

ABSTRACT

Prevention of viral-induced respiratory disease begins with an understanding of the factors that increase or decrease susceptibility to viral infection. The primary receptor for most adenoviruses is the coxsackievirus and adenovirus receptor (CAR), a cell-cell adhesion protein normally localized at the basolateral surface of polarized epithelia and involved in neutrophil transepithelial migration. Recently, an alternate isoform of CAR, CAREx8, has been identified at the apical surface of polarized airway epithelia and is implicated in viral infection from the apical surface. We hypothesized that the endogenous role of CAREx8 may be to facilitate host innate immunity. We show that IL-8, a proinflammatory cytokine and a neutrophil chemoattractant, stimulates the protein expression and apical localization of CAREx8 via activation of AKT/S6K and inhibition of GSK3ß. Apical CAREx8 tethers infiltrating neutrophils at the apical surface of a polarized epithelium. Moreover, neutrophils present on the apical-epithelial surface enhance adenovirus entry into the epithelium. These findings suggest that adenovirus evolved to co-opt an innate immune response pathway that stimulates the expression of its primary receptor, apical CAREx8, to allow the initial infection the intact epithelium. In addition, CAREx8 is a new target for the development of novel therapeutics for both respiratory inflammatory disease and adenoviral infection.


Subject(s)
Adenoviridae Infections/immunology , Adenoviridae , Epithelial Cells/metabolism , Immunity, Innate/immunology , Adenoviridae Infections/metabolism , Animals , Cells, Cultured , Epithelium/metabolism , Humans , Mice , Neutrophils/immunology , Receptors, Virus/metabolism
20.
Cell Signal ; 27(1): 69-81, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25308783

ABSTRACT

The phospholipase D (PLD) superfamily catalyzes the hydrolysis of cell membrane phospholipids generating the key intracellular lipid second messenger phosphatidic acid. However, there is not yet any resolved structure either from a crystallized protein or from NMR of any mammalian PLDs. We propose here a 3D model of the PLD2 by combining homology and ab initio 3 dimensional structural modeling methods, and docking conformation. This model is in agreement with the biochemical and physiological behavior of PLD in cells. For the lipase activity, the N- and C-terminal histidines of the HKD motifs (His 442/His 756) form a catalytic pocket, which accommodates phosphatidylcholine head group (but not phosphatidylethanolamine or phosphatidyl serine). The model explains the mechanism of the reaction catalysis, with nucleophilic attacks of His 442 and water, the latter aided by His 756. Further, the secondary structure regions superimposed with bacterial PLD crystal structure, which indicated an agreement with the model. It also explains protein-protein interactions, such as PLD2-Rac2 transmodulation (with a 1:2 stoichiometry) and PLD2 GEF activity both relevant for cell migration, as well as the existence of binding sites for phosphoinositides such as PIP2. These consist of R236/W238 and R557/W563 and a novel PIP2 binding site in the PH domain of PLD2, specifically R210/R212/W233. In each of these, the polar inositol ring is oriented towards the basic amino acid Arginine. Since tumor-aggravating properties have been found in mice overexpressing PLD2 enzyme, the 3D model of PLD2 will be also useful, to a large extent, in developing pharmaceuticals to modulate its in vivo activity.


Subject(s)
Models, Molecular , Phospholipase D/chemistry , Phospholipase D/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Guanine Nucleotide Exchange Factors/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Phosphatidylcholines/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Sequence Alignment , Software , Substrate Specificity , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...