Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Respir Cell Mol Biol ; 62(3): 331-341, 2020 03.
Article in English | MEDLINE | ID: mdl-31513749

ABSTRACT

Hydrated magnesium silicate (or "talc" particles) is a sclerosis agent commonly used in the management of malignant pleural effusions, a common symptom of metastatic diseases, including lung cancers. However, the direct effects of talc particles to lung carcinoma cells, which can be found in the malignant pleural effusion fluids from patients with lung cancer, are not fully understood. Here, we report a study of the signaling pathways that can modulate the cell death and IL-6 secretion induced by talc particles in human lung carcinoma cells. We found that talc-sensitive cells have higher mRNA and protein expression of PI3K catalytic subunits α and ß. Further experiments confirmed that modulation (inhibition or activation) of the PI3K pathway reduces or enhances cellular sensitivity to talc particles, respectively, independent of the inflammasome. By knocking down specific PI3K isoforms, we also confirmed that both PI3Kα and -ß mediate the observed talc effects. Our results suggest a novel role of the PI3K pathway in talc-induced cell death and IL-6 secretion in lung carcinoma cells. These cellular events are known to drive fibrosis, and thus further studies of the PI3K pathway may provide a better understanding of the mechanisms of talc sclerosis in the malignant pleural space.


Subject(s)
Adenocarcinoma/enzymology , Carcinoma, Non-Small-Cell Lung/enzymology , Class II Phosphatidylinositol 3-Kinases/physiology , Lung Neoplasms/enzymology , Neoplasm Proteins/physiology , Sclerosing Solutions/pharmacology , Talc/pharmacology , Transcription Factors/physiology , Actins/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Death , Cell Line, Tumor , Class II Phosphatidylinositol 3-Kinases/biosynthesis , Class II Phosphatidylinositol 3-Kinases/genetics , Drug Resistance , Enzyme Induction , Humans , Interleukin-6/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/antagonists & inhibitors , Pleural Effusion, Malignant/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Subunits , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/genetics
2.
Mol Cell ; 71(2): 343-351.e4, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30029007

ABSTRACT

Class II phosphoinositide 3-kinases (PI3K-C2) are large multidomain enzymes that control cellular functions ranging from membrane dynamics to cell signaling via synthesis of 3'-phosphorylated phosphoinositides. Activity of the alpha isoform (PI3K-C2α) is associated with endocytosis, angiogenesis, and glucose metabolism. How PI3K-C2α activity is controlled at sites of endocytosis remains largely enigmatic. Here we show that the lipid-binding PX-C2 module unique to class II PI3Ks autoinhibits kinase activity in solution but is essential for full enzymatic activity at PtdIns(4,5)P2-rich membranes. Using HDX-MS, we show that the PX-C2 module folds back onto the kinase domain, inhibiting its basal activity. Destabilization of this intramolecular contact increases PI3K-C2α activity in vitro and in cells, leading to accumulation of its lipid product, increased recruitment of the endocytic effector SNX9, and facilitated endocytosis. Our studies uncover a regulatory mechanism in which coincident binding of phosphoinositide substrate and cofactor selectively activate PI3K-C2α at sites of endocytosis.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/metabolism , Class II Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , C2 Domains/physiology , COS Cells , Chlorocebus aethiops , Class I Phosphatidylinositol 3-Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/physiology , Clathrin/physiology , Endocytosis/physiology , HEK293 Cells , Homeostasis , Humans , Lipids/physiology , Mass Spectrometry , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Protein Domains , Signal Transduction
3.
Oncotarget ; 7(43): 70028-70044, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27661116

ABSTRACT

Retinoblastoma is the most common intraocular malignancy of childhood. Notch plays a key role in retinal cells from which retinoblastomas arise, and we therefore studied the role of Notch signaling in promoting retinoblastoma proliferation. Moderate or strong nuclear expression of Hes1 was found in 10 of 11 human retinoblastoma samples analyzed immunohistochemically, supporting a role for Notch in retinoblastoma growth. Notch pathway components were present in WERI Rb1 and Y79 retinoblastoma lines, with Jag2 and DLL4 more highly expressed than other ligands, and Notch1 and Notch2 more abundant than Notch3. The cleaved/active form of Notch1 was detectable in both lines. Inhibition of the pathway, achieved using a γ-secretase inhibitor (GSI) or by downregulating Jag2, DLL4 or CBF1 using short hairpin RNA, potently reduced growth, proliferation and clonogenicity in both lines. Upregulation of CXCR4 and CXCR7 and downregulation of PI3KC2ß were identified by microarray upon Jag2 suppression. The functional importance of PI3KC2ß was confirmed using shRNA. Synergy was found by combining GSI with Melphalan at their IC50. These findings indicate that Notch pathway is active in WERI Rb1 and Y79, and in most human retinoblastoma samples, and suggest that Notch antagonists may represent a new approach to more effectively treat retinoblastoma.


Subject(s)
Receptors, Notch/antagonists & inhibitors , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/radiation effects , Class II Phosphatidylinositol 3-Kinases/physiology , Cyclic S-Oxides/pharmacology , Humans , Jagged-2 Protein/physiology , Melphalan/pharmacology , Receptors, Notch/physiology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Signal Transduction/physiology , Thiadiazoles/pharmacology
4.
J Clin Invest ; 126(9): 3613-25, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27548528

ABSTRACT

Myotubular myopathy (MTM) is a devastating pediatric neuromuscular disorder of phosphoinositide (PIP) metabolism resulting from mutations of the PIP phosphatase MTM1 for which there are no treatments. We have previously shown phosphatidylinositol-3-phosphate (PI3P) accumulation in animal models of MTM. Here, we tested the hypothesis that lowering PI3P levels may prevent or reverse the MTM disease process. To test this, we targeted class II and III PI3 kinases (PI3Ks) in an MTM1-deficient mouse model. Muscle-specific ablation of Pik3c2b, but not Pik3c3, resulted in complete prevention of the MTM phenotype, and postsymptomatic targeting promoted a striking rescue of disease. We confirmed this genetic interaction in zebrafish, and additionally showed that certain PI3K inhibitors prevented development of the zebrafish mtm phenotype. Finally, the PI3K inhibitor wortmannin improved motor function and prolonged lifespan of the Mtm1-deficient mice. In all, we have identified Pik3c2b as a genetic modifier of Mtm1 mutation and demonstrated that PIK3C2B inhibition is a potential treatment strategy for MTM. In addition, we set the groundwork for similar reciprocal inhibition approaches for treating other PIP metabolic disorders and highlight the importance of modifier gene pathways as therapeutic targets.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/genetics , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Phosphatidylinositol 3-Kinases/genetics , Androstadienes/chemistry , Animals , Animals, Genetically Modified , Class II Phosphatidylinositol 3-Kinases/physiology , Class III Phosphatidylinositol 3-Kinases , Disease Models, Animal , Female , Male , Mice , Mice, Knockout , Motor Skills/drug effects , Myopathies, Structural, Congenital/therapy , Phenotype , Phosphatidylinositol 3-Kinases/physiology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Wortmannin , Zebrafish
5.
J Am Soc Nephrol ; 27(4): 1135-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26271513

ABSTRACT

Signaling from the primary cilium regulates kidney tubule development and cyst formation. However, the mechanism controlling targeting of ciliary components necessary for cilium morphogenesis and signaling is largely unknown. Here, we studied the function of class II phosphoinositide 3-kinase-C2α (PI3K-C2α) in renal tubule-derived inner medullary collecting duct 3 cells and show that PI3K-C2α resides at the recycling endosome compartment in proximity to the primary cilium base. In this subcellular location, PI3K-C2α controlled the activation of Rab8, a key mediator of cargo protein targeting to the primary cilium. Consistently, partial reduction of PI3K-C2α was sufficient to impair elongation of the cilium and the ciliary transport of polycystin-2, as well as to alter proliferation signals linked to polycystin activity. In agreement, heterozygous deletion of PI3K-C2α in mice induced cilium elongation defects in kidney tubules and predisposed animals to cyst development, either in genetic models of polycystin-1/2 reduction or in response to ischemia/reperfusion-induced renal damage. These results indicate that PI3K-C2α is required for the transport of ciliary components such as polycystin-2, and partial loss of this enzyme is sufficient to exacerbate the pathogenesis of cystic kidney disease.


Subject(s)
Cilia/physiology , Class II Phosphatidylinositol 3-Kinases/physiology , Kidney Diseases, Cystic , TRPP Cation Channels/physiology , Animals , Kidney Diseases, Cystic/etiology , Male , Mice , Signal Transduction
6.
Autophagy ; 9(12): 2022-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24165672

ABSTRACT

Phagocytosis and autophagy are two lysosome-mediated cellular degradation pathways designed to eliminate extracellular and intracellular constituents, respectively. Recent studies suggest that these two processes intersect. Several autophagy proteins have been shown to participate in clearance of apoptotic cells, but whether and how the autophagy pathway is involved is unclear. Here we showed that loss of function mutations in 19 genes acting at overlapping or distinct stages of autophagy caused increased numbers of cell corpses in C. elegans embryos. In contrast, genes that mediate specific clearance of P granules or protein aggregates through autophagy are dispensable for cell corpse removal. We showed that defective autophagy impairs phagosome maturation and that autophagy genes act in parallel to the class II phosphoinositide (PI)/phosphatidylinositol (PtdIns) 3-kinase PIKI-1 to regulate phagosomal PtdIns3P in a similar manner as VPS-34. Our data indicate that autophagy may coordinate with PIKI-1 to promote phagosome maturation, thus ensuring efficient clearance of apoptotic cells.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans , Class II Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Organisms, Genetically Modified , Phagosomes/genetics , Phagosomes/metabolism , Signal Transduction/genetics
7.
J Biol Chem ; 288(4): 2325-39, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23192342

ABSTRACT

The phosphatidylinositol (PtdIns) 3-kinase (PI3K) family regulates diverse cellular processes, including cell proliferation, migration, and vesicular trafficking, through catalyzing 3'-phosphorylation of phosphoinositides. In contrast to class I PI3Ks, including p110α and p110ß, functional roles of class II PI3Ks, comprising PI3K-C2α, PI3K-C2ß, and PI3K-C2γ, are little understood. The lysophospholipid mediator sphingosine 1-phosphate (S1P) plays the important roles in regulating vascular functions, including vascular formation and barrier integrity, via the G-protein-coupled receptors S1P(1-3). We studied the roles of PI3K-C2α in S1P-induced endothelial cell (EC) migration and tube formation. S1P stimulated cell migration and activation of Akt, ERK, and Rac1, the latter of which acts as a signaling molecule essential for cell migration and tube formation, via S1P(1) in ECs. Knockdown of either PI3K-C2α or class I p110ß markedly inhibited S1P-induced migration, lamellipodium formation, and tube formation, whereas that of p110α or Vps34 did not. Only p110ß was necessary for S1P-iduced Akt activation, but both PI3K-C2α and p110ß were required for Rac1 activation. FRET imaging showed that S1P induced Rac1 activation in both the plasma membrane and PtdIns 3-phosphate (PtdIns(3)P)-enriched endosomes. Knockdown of PI3K-C2α but not p110ß markedly reduced PtdIns(3)P-enriched endosomes and suppressed endosomal Rac1 activation. Also, knockdown of PI3K-C2α but not p110ß suppressed S1P-induced S1P(1) internalization into PtdIns(3)P-enriched endosomes. Finally, pharmacological inhibition of endocytosis suppressed S1P-induced S1P(1) internalization, Rac1 activation, migration, and tube formation. These observations indicate that PI3K-C2α plays the crucial role in S1P(1) internalization into the intracellular vesicular compartment, Rac1 activation on endosomes, and thereby migration through regulating vesicular trafficking in ECs.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/physiology , Gene Expression Regulation, Enzymologic , Receptors, Lysosphingolipid/genetics , Cell Movement , Cells, Cultured , Class II Phosphatidylinositol 3-Kinases/genetics , Endocytosis , Endosomes/metabolism , Endothelial Cells/cytology , Fluorescence Resonance Energy Transfer , Human Umbilical Vein Endothelial Cells , Humans , Lysophospholipids/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Transfection , rac GTP-Binding Proteins/metabolism
8.
Neuroscience ; 169(1): 449-54, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20451587

ABSTRACT

G protein-coupled opioid receptors undergo desensitization after prolonged agonist exposure. Recent in vitro studies of mu-opioid receptor (MOR) signaling revealed an involvement of phosphoinositide 3-kinases (PI3K) in agonist-induced MOR desensitization. Here we document a specific role of the G protein-coupled class IB isoform PI3Kgamma in MOR desensitization in mice and isolated sensory neurons. The tail-withdrawal nociception assay evidenced a compromised morphine-induced tolerance of PI3Kgamma-deficient mice compared to wild-type animals. Consistent with a role of PI3Kgamma in MOR signaling, PI3Kgamma was expressed in a subgroup of small-diameter dorsal root ganglia (DRG) along with MOR and the transient receptor potential vanilloid type 1 (TRPV1) receptor. In isolated DRG acute stimulation of MOR blocked voltage-gated calcium currents (VGCC) in both wild-type and PI3Kgamma-deficient DRG neurons. By contrast, following long-term opioid administration the attenuating effect of MOR was strongly compromised in wild-type DRG but not in PI3Kgamma-deficient DRG. Our results uncover PI3Kgamma as an essential modulator of long-term MOR desensitization and tolerance development induced by chronic opioid treatment in sensory neurons.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/physiology , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Opioid, mu/drug effects , Sensory Receptor Cells/enzymology , Animals , Calcium Channels/physiology , Cells, Cultured/enzymology , Cells, Cultured/physiology , Class II Phosphatidylinositol 3-Kinases/deficiency , Class II Phosphatidylinositol 3-Kinases/genetics , Drug Tolerance/physiology , Ganglia, Spinal/cytology , Mice , Mice, Knockout , Morphine/administration & dosage , Morphine/therapeutic use , Narcotics/administration & dosage , Narcotics/therapeutic use , Nociceptors/drug effects , Nociceptors/physiology , Patch-Clamp Techniques , Rats , Rats, Wistar , Reaction Time/drug effects , Recombinant Fusion Proteins/physiology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...