Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Inflam ; 2013: 980327, 2013.
Article in English | MEDLINE | ID: mdl-23710416

ABSTRACT

Intraplaque hemorrhage causes adaptive remodelling of macrophages towards a protective phenotype specialized towards handling iron and lipid overload, denoted Mhem. The Mhem phenotype expresses elevated levels of hemoglobin (Hb) scavenger receptor, CD163, capable of endocytosing pro-oxidant free Hb complexed to acute phase protein haptoglobin (Hp). It is notable that individuals homozygous for the Hp 2 allele (a poorer antioxidant) are at increased risk of cardiovascular disease compared to the Hp 1 allele. In this study, we examined whether scavenging of polymorphic Hp:Hb complexes differentially generated downstream anti-inflammatory signals in cultured human macrophages culminating in interleukin (IL)-10 secretion. We describe an anti-inflammatory signalling pathway involving phosphatidylinositol-3-kinase activation upstream of Akt phosphorylation (pSer473Akt) and IL-10 secretion. The pathway is mediated specifically through CD163 and is blocked by anti-CD163 antibody or phagocytosis inhibitor. However, levels of pSer473Akt and IL-10 were significantly diminished when scavenging polymorphic Hp2-2:Hb complexes compared to Hp1-1:Hb complexes (P < 0.05). Impaired anti-inflammatory macrophage signaling through a CD163/pAkt/IL-10 axis may thus represent a possible Hp2-2 disease mechanism in atherosclerosis.

2.
Diabetes ; 61(9): 2280-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22586581

ABSTRACT

CCN2, a secreted profibrotic protein, is highly expressed in diabetic nephropathy (DN) and implicated in its pathogenesis; however, the actions of CCN2 in DN remain elusive. We previously demonstrated that CCN2 triggers signaling via tropomyosin receptor kinase A (TrkA). Trace expression of TrkA is found in normal kidneys, but its expression is elevated in several nephropathies; yet its role in DN is unexplored. In this study we show de novo expression of TrkA in human and murine DN. We go on to study the molecular mechanisms leading to TrkA activation and show that it involves hypoxia, as demonstrated by ischemia-reperfusion injury and in vitro experiments mimicking hypoxia, implicating hypoxia as a common pathway leading to disease. We also expose renal cells to hyperglycemia, which led to TrkA phosphorylation in mesangial cells, tubular epithelial cells, and podocytes but not in glomerular endothelial cells and renal fibroblasts. In addition, we report that hyperglycemia caused an induction of phosphorylated extracellular signal-related kinase 1/2 and Snail1 that was abrogated by silencing of TrkA or CCN2 using small interfering RNA. In conclusion, we provide novel evidence that TrkA is activated in diabetic kidneys and suggest that anti-TrkA therapy may prove beneficial in DN.


Subject(s)
Connective Tissue Growth Factor/physiology , Diabetic Nephropathies/etiology , Hyperglycemia/complications , Animals , Connective Tissue Growth Factor/genetics , Diabetic Nephropathies/physiopathology , Humans , Hyperglycemia/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Kidney/pathology , MAP Kinase Signaling System/physiology , Mesangial Cells/metabolism , Mice , Phosphorylation , RNA, Small Interfering/pharmacology , Receptor, trkA/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/biosynthesis
3.
Mol Cell Biol ; 31(1): 63-80, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974805

ABSTRACT

An early lesion in many kidney diseases is damage to podocytes, which are critical components of the glomerular filtration barrier. A number of proteins are essential for podocyte filtration function, but the signaling events contributing to development of nephrotic syndrome are not well defined. Here we show that class II phosphoinositide 3-kinase C2α (PI3KC2α) is expressed in podocytes and plays a critical role in maintaining normal renal homeostasis. PI3KC2α-deficient mice developed chronic renal failure and exhibited a range of kidney lesions, including glomerular crescent formation and renal tubule defects in early disease, which progressed to diffuse mesangial sclerosis, with reduced podocytes, widespread effacement of foot processes, and modest proteinuria. These findings were associated with altered expression of nephrin, synaptopodin, WT-1, and desmin, indicating that PI3KC2α deficiency specifically impacts podocyte morphology and function. Deposition of glomerular IgA was observed in knockout mice; importantly, however, the development of severe glomerulonephropathy preceded IgA production, indicating that nephropathy was not directly IgA mediated. PI3KC2α deficiency did not affect immune responses, and bone marrow transplantation studies also indicated that the glomerulonephropathy was not the direct consequence of an immune-mediated disease. Thus, PI3KC2α is critical for maintenance of normal glomerular structure and function by supporting normal podocyte function.


Subject(s)
Kidney Glomerulus/anatomy & histology , Kidney Glomerulus/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Antigens, Surface/metabolism , Bone Marrow Transplantation , Glomerulonephritis/etiology , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Glomerulonephritis, IGA/etiology , Glomerulonephritis, IGA/pathology , Glomerulonephritis, IGA/physiopathology , Humans , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Kidney Glomerulus/enzymology , Kidney Glomerulus/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Phosphatidylinositol 3-Kinases/deficiency , Phosphatidylinositol 3-Kinases/genetics , Podocytes/enzymology , Podocytes/pathology , Podocytes/physiology , Renal Insufficiency/etiology , Renal Insufficiency/pathology , Renal Insufficiency/physiopathology , Transplantation Chimera
4.
J Cell Biol ; 190(3): 307-15, 2010 Aug 09.
Article in English | MEDLINE | ID: mdl-20679431

ABSTRACT

Phosphatidylinositol (PI) 4,5-bisphosphate (PI(4,5)P(2)) and its phosphorylated product PI 3,4,5-triphosphate (PI(3,4,5)P(3)) are two major phosphoinositides concentrated at the plasma membrane. Their levels, which are tightly controlled by kinases, phospholipases, and phosphatases, regulate a variety of cellular functions, including clathrin-mediated endocytosis and receptor signaling. In this study, we show that the inositol 5-phosphatase SHIP2, a negative regulator of PI(3,4,5)P(3)-dependent signaling, also negatively regulates PI(4,5)P(2) levels and is concentrated at endocytic clathrin-coated pits (CCPs) via interactions with the scaffold protein intersectin. SHIP2 is recruited early at the pits and dissociates before fission. Both knockdown of SHIP2 expression and acute production of PI(3,4,5)P(3) shorten CCP lifetime by enhancing the rate of pit maturation, which is consistent with a positive role of both SHIP2 substrates, PI(4,5)P(2) and PI(3,4,5)P(3), on coat assembly. Because SHIP2 is a negative regulator of insulin signaling, our findings suggest the importance of the phosphoinositide metabolism at CCPs in the regulation of insulin signal output.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Endocytosis , Phosphoric Monoester Hydrolases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Inositol Polyphosphate 5-Phosphatases , Mice , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases
5.
J Clin Invest ; 120(5): 1469-78, 2010 May.
Article in English | MEDLINE | ID: mdl-20407205

ABSTRACT

Crescentic glomerulonephritis (CGN), which frequently results in acute and chronic kidney disease, is characterized by and dependent on glomerular infiltration by macrophages. The mannose receptor (MR) is a pattern recognition receptor implicated in the uptake of endogenous and microbial ligands by macrophages, mesangial cells (MCs), and selected endothelial cells. It is upregulated on alternatively activated macrophages (i.e., macrophages associated with tissue repair and humoral immunity) and involved in antigen presentation to T cells. We used the mouse model of nephrotoxic nephritis to investigate the role of MR in CGN. Our results demonstrate what we believe to be a novel role for MR in the promotion of CGN that is independent of adaptive immune responses. MR-deficient (Mr-/-) mice were protected from CGN despite generating humoral and T cell responses similar to those of WT mice, but they demonstrated diminished macrophage and MC Fc receptor-mediated (FcR-mediated) functions, including phagocytosis and Fc-mediated oxygen burst activity. Mr-/- MCs demonstrated augmented apoptosis compared with WT cells, and this was associated with diminished Akt phosphorylation. Macrophage interaction with apoptotic MCs induced a noninflammatory phenotype that was more marked in Mr-/- macrophages than in WT macrophages. Our results demonstrate that MR augments Fc-mediated function and promotes MC survival. We suggest that targeting MR may provide an alternative therapeutic approach in CGN while minimizing the impact on adaptive immune responses, which are affected by conventional immunosuppressive approaches.


Subject(s)
Gene Expression Regulation , Glomerulonephritis/metabolism , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Fc/metabolism , Animals , Apoptosis , Female , Macrophages/metabolism , Male , Mannose Receptor , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Oxygen/metabolism , Phagocytosis , Phosphorylation , T-Lymphocytes/metabolism
6.
FASEB J ; 24(6): 1824-37, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20061534

ABSTRACT

Phosphatidylinositide 3-kinases (PI3Ks) play central roles in insulin signal transduction. While the contribution of class Ia PI3K members has been extensively studied, the role of class II members remains poorly understood. The diverse actions of class II PI3K-C2alpha have been attributed to its lipid product PI(3)P. By applying pharmacological inhibitors, transient overexpression and small-interfering RNA-based knockdown of PI3K and PKB/Akt isoforms, together with PI-lipid profiling and live-cell confocal and total internal reflection fluorescence microscopy, we now demonstrate that in response to insulin, PI3K-C2alpha generates PI(3,4)P(2), which allows the selective activation of PKBalpha/Akt1. Knockdown of PI3K-C2alpha expression and subsequent reduction of PKBalpha/Akt1 activity in the pancreatic beta-cell impaired glucose-stimulated insulin release, at least in part, due to reduced glucokinase expression and increased AS160 activity. Hence, our results identify signal transduction via PI3K-C2alpha as a novel pathway whereby insulin activates PKB/Akt and thus discloses PI3K-C2alpha as a potential drugable target in type 2 diabetes. The high degree of codistribution of PI3K-C2alpha and PKBalpha/Akt1 with insulin receptor B type, but not A type, in the same plasma membrane microdomains lends further support to the concept that selectivity in insulin signaling is achieved by the spatial segregation of signaling events.


Subject(s)
Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sweetening Agents/pharmacology , Androstadienes/pharmacology , Animals , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Class II Phosphatidylinositol 3-Kinases , Fluorescent Antibody Technique , Glucokinase/metabolism , Immunoprecipitation , Insulin Antagonists/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Lipids , Mice , Mice, Obese , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptor, Insulin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Wortmannin
7.
ChemMedChem ; 5(1): 130-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19950162

ABSTRACT

Imatinib is a clinically important ATP analogue inhibitor that targets the tyrosine kinase domain of the intracellular Abl kinase and the PDGF receptor family. Imatinib has revolutionised the treatment of chronic myeloid leukaemia, which is caused by the oncogene Bcr-Abl and certain solid tumours that harbor oncogenic mutations of the PDGF receptor family. As a leading kinase inhibitor, imatinib also provides an excellent model system to investigate how changes in drug design impact biological activity, which is an important consideration for rational drug design. Herein we report a new series of imatinib derivatives that in general have greater activity against the family of PDGF receptors and poorer activity against Abl, as a result of modifications of the phenyl and N-methylpiperazine rings. These new compounds provide a platform for further drug development against the therapeutically important PDGF receptor family and they also provide insight into the engineering of drugs with altered biological activity.


Subject(s)
Antineoplastic Agents/chemistry , Fusion Proteins, bcr-abl/metabolism , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Benzamides , Binding Sites , Cell Line, Tumor , Computer Simulation , Drug Design , Humans , Imatinib Mesylate , K562 Cells , Mice , Phosphorylation , Piperazines/chemical synthesis , Piperazines/toxicity , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/toxicity , Pyrimidines/chemical synthesis , Pyrimidines/toxicity , Substrate Specificity
8.
Biochem J ; 422(1): 53-60, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19496756

ABSTRACT

Although the class II phosphoinositide 3-kinase enzymes PI3K-C2alpha and PI3K-C2beta act acutely downstream of cell surface receptors they have also been localized to nuclei in mammalian cells. As with the class I PI3K enzymes, the relationship between the pools of enzyme present in cytoplasm and nuclei remains poorly understood. In this study we test the hypothesis that PI3K-C2beta translocates to nuclei in response to growth factor stimulation. Fractionating homogenates of quiescent cells revealed that less than 5% of total PI3K-C2beta resides in nuclei. Stimulation with epidermal growth factor sequentially increased levels of this enzyme, firstly in the cytosol and secondly in the nuclei. Using detergent-treated nuclei, we showed that PI3K-C2beta co-localized with lamin A/C in the nuclear matrix. This was confirmed biochemically, and a phosphoinositide kinase assay showed a statistically significant increase in nuclear PI3K-C2beta levels and lipid kinase activity following epidermal growth factor stimulation. C-terminal deletion and point mutations of PI3K-C2beta demonstrated that epidermal growth factor-driven translocation to the nucleus is dependent on a sequence of basic amino acid residues (KxKxK) that form a nuclear localization motif within the C-terminal C2 domain. Furthermore, when this sequence was expressed as an EGFP (enhanced green fluorescent protein) fusion protein, it translocated fluorescence into nuclei with an efficiency dependent upon copy number. These data demonstrate that epidermal growth factor stimulates the appearance of PI3K-C2beta in nuclei. Further, this effect is dependent on a nuclear localization signal present within the C-terminal C2 domain, indicating its bimodal function regulating phospholipid binding and shuttling PI3K-C2beta into the nucleus.


Subject(s)
Cell Nucleus/drug effects , Cell Nucleus/enzymology , Epidermal Growth Factor/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Sequence , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Class II Phosphatidylinositol 3-Kinases , Cytosol/drug effects , Cytosol/enzymology , Green Fluorescent Proteins , Humans , Lamins/metabolism , Models, Biological , Molecular Sequence Data , Nuclear Localization Signals/metabolism , Nuclear Matrix/drug effects , Nuclear Matrix/enzymology , Phosphatidylinositol 3-Kinases/chemistry , Protein Transport/drug effects
9.
Mol Biol Cell ; 19(12): 5593-603, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18843041

ABSTRACT

Phosphatidylinositol-3-phosphate [PtdIns(3)P] is a key player in early endosomal trafficking and is mainly produced by class III phosphatidylinositol 3-kinase (PI3K). In neurosecretory cells, class II PI3K-C2alpha and its lipid product PtdIns(3)P have recently been shown to play a critical role during neuroexocytosis, suggesting that two distinct pools of PtdIns(3)P might coexist in these cells. However, the precise characterization of this additional pool of PtdIns(3)P remains to be established. Using a selective PtdIns(3)P probe, we have identified a novel PtdIns(3)P-positive pool localized on secretory vesicles, sensitive to PI3K-C2alpha knockdown and relatively resistant to wortmannin treatment. In neurosecretory cells, stimulation of exocytosis promoted a transient albeit large increase in PtdIns(3)P production localized on secretory vesicles sensitive to PI3K-C2alpha knockdown and expression of PI3K-C2alpha catalytically inactive mutant. Using purified chromaffin granules, we found that PtdIns(3)P production is controlled by Ca(2+). We confirmed that PtdIns(3)P production from recombinantly expressed PI3K-C2alpha is indeed regulated by Ca(2+). We provide evidence that a dynamic pool of PtdIns(3)P synthesized by PI3K-C2alpha occurs on secretory vesicles in neurosecretory cells, demonstrating that the activity of a member of the PI3K family is regulated by Ca(2+) in vitro and in living neurosecretory cells.


Subject(s)
Calcium/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Secretory Vesicles/metabolism , Androstadienes/metabolism , Animals , Cattle , Cell Line , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Class II Phosphatidylinositol 3-Kinases , Exocytosis/physiology , Humans , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase Inhibitors/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Wortmannin
10.
Neoplasia ; 10(9): 949-53, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18714395

ABSTRACT

One limitation of current biochemical or histologic analysis of advanced prostate cancer (PC; T(3)/T(4) +/- N(x) M(x)) is the ability to identify on first diagnostic biopsy patients who will make a durable response to hormone ablation therapy. The aim of this study was to assess the predictive value (sustained response to hormonal therapy and clinical outcome (relapse-free and overall survival)) of phosphatase and tensin homolog (PTEN) and the androgen receptor (AR) immunoexpression in the presenting biopsy. Analysis was performed on 47 samples (10 cases of benign prostatic hyperplasia and 37 hormone-naive PCs). Patients selected represented two stages in the natural history of PC: The "clinical metastatic androgen-responsive" (androgen-dependent PC, ADPC) and the "clinical metastatic androgen-resistant" (androgen-independent PC, AIPC). Reduced immunoreactivity (IR) of either or both PTEN/AR in the initial hormone-naive PC samples was observed with increased frequency in AIPCs. In the ADPC group, low PTEN and/or AR-IR was associated with a shorter median relapse-free survival, i.e., at 30 months after surgery, the probability of relapse-free survival for high expressors of PTEN and AR was 85.7% (SEM = 9.3) compared with only 16.6% (SEM = 15.2) in low expressors. At 36 months, only 28.5% (SEM = 9.3) of ADPC high expressors had experienced a biochemical relapse compared with 100% of low expressors (hazard ratio, 4.6; 95% confidence interval, 4.7-146.8). Further studies analyzing the coexpression of PTEN and AR should be undertaken to validate this pilot study and the utility of these biomarkers in routine histopathologic workup of patients with PC.


Subject(s)
Biomarkers, Tumor/metabolism , PTEN Phosphohydrolase/metabolism , Prostatic Hyperplasia/genetics , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Aged , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Pilot Projects , Predictive Value of Tests , Recurrence , Retrospective Studies
11.
Nat Genet ; 40(5): 553-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18443593

ABSTRACT

Crescentic glomerulonephritis is an important cause of human kidney failure for which the underlying molecular basis is largely unknown. In previous studies, we mapped several susceptibility loci, Crgn1-Crgn7, for crescentic glomerulonephritis in the Wistar Kyoto (WKY) rat. Here we show by combined congenic, linkage and microarray studies that the activator protein-1 (AP-1) transcription factor JunD is a major determinant of macrophage activity and is associated with glomerulonephritis susceptibility. Introgression of Crgn2 from the nonsusceptible Lewis strain onto the WKY background leads to significant reductions in crescent formation, macrophage infiltration, Fc receptor-mediated macrophage activation and cytokine production. Haplotype analysis restricted the Crgn2 linkage interval to a 430-kb interval containing Jund, which is markedly overexpressed in WKY macrophages and glomeruli. Jund knockdown in rat and human primary macrophages led to significantly reduced macrophage activity and cytokine secretion, indicating conservation of JunD function in macrophage activation in rats and humans and suggesting in vivo inhibition of Jund as a possible new therapeutic strategy for diseases characterized by inflammation and macrophage activation.


Subject(s)
Genetic Predisposition to Disease , Glomerulonephritis/genetics , Macrophage Activation/genetics , Proto-Oncogene Proteins c-jun/physiology , Rats/genetics , Transcription Factor AP-1/physiology , Animals , Animals, Congenic , Chromosome Mapping , Gene Expression , Genetic Linkage , Haplotypes , Humans , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Rats, Inbred Lew , Rats, Inbred WKY , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
12.
Am J Pathol ; 171(5): 1462-73, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17982125

ABSTRACT

Pancreatic secretory trypsin inhibitor (PSTI) is a serine protease inhibitor, expressed in gut mucosa, whose function is unclear. We, therefore, examined the effects of PSTI on gut stability and repair. Transgenic mice overexpressing human PSTI within the jejunum (FABPi(-1178 to +28) hPSTI construct) showed no change in baseline morphology or morphometry but reduced indomethacin-induced injury in overexpressing hPSTI region by 42% (P < 0.01). Systemic recombinant hPSTI did not affect baseline morphology or morphometry but truncated injurious effects in prevention and recovery rat models of dextran-sodium-sulfate-induced colitis. In vitro studies showed PSTI stimulated cell migration but not proliferation of human colonic carcinoma HT29 or immortalized mouse colonic YAMC cells. PSTI also induced changes in vectorial ion transport (short-circuit current) when added to basolateral but not apical surfaces of polarized monolayers of Colony-29 cells. Restitution and vectorial ion transport effects of PSTI were dependent on the presence of a functioning epidermal growth factor (EGF) receptor because cells with a disrupted (EGFR(-/-) immortalized cells) or neutralized (EGFR blocking antibodies or tyrosine kinase inhibitor) receptor prevented these effects. PSTI also reduced the cytokine release of lipopolysaccharide-stimulated dendritic cells. We conclude that administration of PSTI may provide a novel method of stabilizing intestinal mucosa against noxious agents and stimulating repair after injury.


Subject(s)
Carrier Proteins/pharmacology , Intestinal Mucosa/drug effects , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line , Cell Movement , Cell Proliferation , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Dendritic Cells/physiology , Dextran Sulfate , ErbB Receptors/metabolism , Fatty Acid-Binding Proteins/genetics , Humans , Indomethacin , Intestinal Mucosa/physiology , Ion Transport , Jejunal Diseases/chemically induced , Jejunal Diseases/metabolism , Jejunal Diseases/pathology , Male , Mice , Mice, Transgenic , Phosphorylation , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Trypsin Inhibitor, Kazal Pancreatic
13.
Mol Cell Biol ; 27(22): 7906-17, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17875942

ABSTRACT

While endocytosis attenuates signals from plasma membrane receptors, recent studies suggest that endocytosis also serves as a platform for the compartmentalized activation of cellular signaling pathways. Intersectin (ITSN) is a multidomain scaffolding protein that regulates endocytosis and has the potential to regulate various biochemical pathways through its multiple, modular domains. To address the biological importance of ITSN in regulating cellular signaling pathways versus in endocytosis, we have stably silenced ITSN expression in neuronal cells by using short hairpin RNAs. Decreasing ITSN expression dramatically increased apoptosis in both neuroblastoma cells and primary cortical neurons. Surprisingly, the loss of ITSN did not lead to major defects in the endocytic pathway. Yeast two-hybrid analysis identified class II phosphoinositide 3'-kinase C2beta (PI3K-C2beta) as an ITSN binding protein, suggesting that ITSN may regulate a PI3K-C2beta-AKT survival pathway. ITSN associated with PI3K-C2beta on a subset of endomembrane vesicles and enhanced both basal and growth factor-stimulated PI3K-C2beta activity, resulting in AKT activation. The use of pharmacological inhibitors, dominant negatives, and rescue experiments revealed that PI3K-C2beta and AKT were epistatic to ITSN. This study represents the first demonstration that ITSN, independent of its role in endocytosis, regulates a critical cellular signaling pathway necessary for cell survival.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Cell Survival , Neurons/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Subunits/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Adaptor Proteins, Vesicular Transport/genetics , Amino Acid Sequence , Animals , Cell Line , Endocytosis/physiology , Enzyme Activation , Epidermal Growth Factor/metabolism , Epistasis, Genetic , Humans , Mice , Molecular Sequence Data , Neurons/cytology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Subunits/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Two-Hybrid System Techniques
14.
Nephron Physiol ; 107(2): p45-56, 2007.
Article in English | MEDLINE | ID: mdl-17804914

ABSTRACT

BACKGROUND: Platelet-derived growth factor (PDGF) is a potent activator of mesangial cell proliferation and migration. Although phosphoinositide 3-kinase (PI3K) enzymes are important downstream targets of the PDGF receptor, the contribution made by their 3-phosphoinositide products in the reorganization of actin cytoskeleton and focal adhesions has been questioned. METHODS AND RESULTS: Pharmacological inhibition of the PI3K activity blocks PDGF-induced migration of human primary mesangial cells using an in vitro scrape wound healing assay. Acute (<10 min) inhibition of the PI3K activity did not alter the effect of PDGF on either stress fibre dissolution or reorganization of focal adhesions. However, at later times (>30 min), PDGF-stimulated responses were inhibited. In contrast, PDGF-stimulated membrane ruffling remained insensitive to PI3K inhibitors throughout. Inhibition of protein kinase C and Erk also attenuated PDGF-stimulated mesangial cell migration; however, neither signaling pathway was responsible for the initial effects on filamentous actin and focal adhesions. CONCLUSIONS: We propose that following PDGF stimulation of mesangial cells, reorganization of the actin cytoskeleton occurs in a biphasic manner. The mechanism responsible for mesangial cell migration that occurs immediately following PDGF stimulation may serve to 'prime' for the subsequent 3-phosphoinositide-, protein-kinase-C-, and Erk-dependent migration.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Mesangial Cells/metabolism , Phosphatidylinositol Phosphates/physiology , Platelet-Derived Growth Factor/physiology , 3-Phosphoinositide-Dependent Protein Kinases , Cell Movement/physiology , Cells, Cultured , Cytoskeleton/enzymology , Humans , Mesangial Cells/cytology , Mesangial Cells/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism
15.
Nature ; 439(7078): 851-5, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16482158

ABSTRACT

Identification of the genes underlying complex phenotypes and the definition of the evolutionary forces that have shaped eukaryotic genomes are among the current challenges in molecular genetics. Variation in gene copy number is increasingly recognized as a source of inter-individual differences in genome sequence and has been proposed as a driving force for genome evolution and phenotypic variation. Here we show that copy number variation of the orthologous rat and human Fcgr3 genes is a determinant of susceptibility to immunologically mediated glomerulonephritis. Positional cloning identified loss of the newly described, rat-specific Fcgr3 paralogue, Fcgr3-related sequence (Fcgr3-rs), as a determinant of macrophage overactivity and glomerulonephritis in Wistar Kyoto rats. In humans, low copy number of FCGR3B, an orthologue of rat Fcgr3, was associated with glomerulonephritis in the autoimmune disease systemic lupus erythematosus. The finding that gene copy number polymorphism predisposes to immunologically mediated renal disease in two mammalian species provides direct evidence for the importance of genome plasticity in the evolution of genetically complex phenotypes, including susceptibility to common human disease.


Subject(s)
Antigens, CD/genetics , Gene Dosage/genetics , Genetic Predisposition to Disease/genetics , Lupus Nephritis/genetics , Polymorphism, Genetic/genetics , Receptors, IgG/genetics , Animals , Base Sequence , Exons/genetics , GPI-Linked Proteins , Gene Duplication , Haplotypes , Humans , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Molecular Sequence Data , Rats , Rats, Inbred WKY , Sequence Deletion/genetics
16.
J Cell Physiol ; 206(3): 586-93, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16222711

ABSTRACT

The class II phosphoinositide 3-kinase (PI3K)-C2beta is recruited to polypeptide growth factor receptors following ligand stimulation. In contrast to the class I A p85/p110 heterodimers, this interaction is dependent upon proline residues present within the N-terminal sequence of the 3-phosphoinositide kinase. However, the mechanism by which PI3K-C2beta catalytic activity is regulated currently remains unknown. In many tumours, increased expression of ErbB receptors confers a poor prognosis. We demonstrate that increased expression of EGFR enhanced its association with PI3K-C2beta following stimulation with EGF. Deletion of the first proline rich region within the N-terminus precluded recruitment of PI3K-C2beta to activated EGFR. Although deletion of the first proline rich motif also rendered the enzyme catalytically inactive, further deletions (residues 1-148 and 1-261) that removed the second and third proline rich motifs increased kinase activity. These data confirm a regulatory role for the N-terminus of class II PI3K enzymes suggesting that catalytic activity is regulated by factors that associate with this region during recruitment to activated growth factor receptors. Using an N-terminal PI3K-C2beta-GST fusion protein, clathrin heavy chain was affinity purified from A431 cell lysates. Association of PI3K-C2beta with clathrin was confirmed by co-immunoprecipitation from cell lysates while intracellular co-localisation of PI3K-C2beta and clathrin was confirmed by confocal microscopy. Our findings demonstrate for the first time that the PI3K-C2beta isoform associates with clathrin and thus provides a link between receptor mediated intracellular signalling and clathrin coated vesicle transport.


Subject(s)
Clathrin/metabolism , ErbB Receptors/metabolism , Phosphatidylinositol 3-Kinases/physiology , Binding Sites , Cell Line , Chromatography, Affinity , Class II Phosphatidylinositol 3-Kinases , GRB2 Adaptor Protein/metabolism , Humans , Lipids , Phosphorylation , Phosphotransferases , Tumor Cells, Cultured
17.
Mol Biol Cell ; 16(10): 4841-51, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16055506

ABSTRACT

Neurotransmitter release and hormonal secretion are highly regulated processes culminating in the calcium-dependent fusion of secretory vesicles with the plasma membrane. Here, we have identified a role for phosphatidylinositol 3-kinase C2alpha (PI3K-C2alpha) and its main catalytic product, PtdIns3P, in regulated exocytosis. In neuroendocrine cells, PI3K-C2alpha is present on a subpopulation of mature secretory granules. Impairment of PI3K-C2alpha function specifically inhibits the ATP-dependent priming phase of exocytosis. Overexpression of wild-type PI3K-C2alpha enhanced secretion, whereas transfection of PC12 cells with a catalytically inactive PI3K-C2alpha mutant or a 2xFYVE domain sequestering PtdIns3P abolished secretion. Based on these results, we propose that production of PtdIns3P by PI3K-C2alpha is required for acquisition of fusion competence in neurosecretion.


Subject(s)
Adenosine Triphosphate/metabolism , Exocytosis/physiology , Neurosecretion/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Secretory Vesicles/physiology , Adrenal Glands/cytology , Animals , Catecholamines/metabolism , Cattle , Cells, Cultured , Chromaffin Cells/physiology , Class II Phosphatidylinositol 3-Kinases , Human Growth Hormone/metabolism , Humans , Mutation , Phosphatidylinositol 3-Kinases/genetics , Rats
18.
J Cell Physiol ; 205(3): 452-62, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16113997

ABSTRACT

The biological and pathophysiological significance of class II phosphoinositide 3-kinase enzyme expression currently remains unclear. Using an in vitro scrape wound assay and time-lapse video microscopy, we demonstrate that cell motility is increased in cultures expressing recombinant PI3K-C2beta enzyme. In addition, overexpression of PI3K-C2beta transiently decreased cell adhesion, stimulated the formation of cytoplasmic processes, and decreased the rate of cell proliferation. Consistent with these observations, expression of PI3K-C2beta also decreased expression of alpha4 beta1 integrin subunits. Using asynchronous cultures, we show that endogenous PI3K-C2beta is present in lamellipodia of motile cells. When cells expressing recombinant PI3K-C2beta were plated onto fibronectin, cortical actin staining increased markedly and actin rich lamellipodia and filopodia became evident. Overexpression of a 2xFYVE(Hrs) domain fusion protein abolished this response demonstrating that the effect of PI3K-C2beta on the reorganization of actin filaments is dependent upon PtdIns3P. Finally, overexpression of PI3K-C2beta increased GTP loading of Cdc42. Our data demonstrates for the first time, that PI3K-C2beta plays a regulatory role in cell motility and that the mechanism by which it reorganizes the actin cytoskeleton is dependent upon PtdIns3P production.


Subject(s)
Cell Movement/drug effects , Cell Movement/physiology , Phosphatidylinositol 3-Kinases/pharmacology , Phosphatidylinositol Phosphates/physiology , Actins/physiology , Animals , Cattle/blood , Cell Adhesion/drug effects , Cell Line , Cell Proliferation/drug effects , Cells/ultrastructure , Class II Phosphatidylinositol 3-Kinases , Cytoskeleton/drug effects , Fetal Blood , Guanosine Triphosphate/metabolism , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/pharmacology , Tissue Distribution , cdc42 GTP-Binding Protein/blood , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism
19.
Neoplasia ; 6(6): 846-53, 2004.
Article in English | MEDLINE | ID: mdl-15720812

ABSTRACT

Despite multiple reports of overexpression in prostate cancer (PC), the reliance of PC cells on activated epidermal growth factor receptor (EGFR) and its downstream signaling to phosphoinositide 3'-kinase/Akt (PI3K/Akt/PTEN) and/or mitogen-activated protein kinase (MAPK/ERK) pathways has not been fully elucidated. In this study, we compared the role of EGF-mediated signaling in nonmalignant (BPH-1, PNT1A, and PNT1B) and PC cell lines (DU145, PC3, LNCaP, and CWR22Rv1). EGF-induced proliferation was observed in all EGFR-expressing PC cells except PC3, indicating that EGFR expression does not unequivocally trigger proliferation following EGF stimulation. ErbB2 recruitment potentiated EGF-induced signals and was associated with the most pronounced effects of EGF despite low EGFR expression. In this way, the sum of EGFR and ErbB2 receptor phosphorylation proved to be a more sensitive indicator of EGF-induced proliferation than quantification of the expression of either receptor alone. Both Akt and ERK were rapidly phosphorylated in response to EGF, with ERK phosphorylation being the weakest in PC3 cells. Extrapolation of these findings to clinical PC suggests that assessment of phosphorylated EGFR + ErbB2 together could serve as a marker for sensitivity to anti-EGFR-targeted therapies.


Subject(s)
Cell Proliferation/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Prostatic Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Blotting, Western , Cell Line, Tumor , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Male , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt
20.
BMC Clin Pathol ; 3(1): 4, 2003 Oct 16.
Article in English | MEDLINE | ID: mdl-14563213

ABSTRACT

BACKGROUND: Growth factor, cytokine and chemokine-induced activation of PI3K enzymes constitutes the start of a complex signalling cascade, which ultimately mediates cellular activities such as proliferation, differentiation, chemotaxis, survival, trafficking, and glucose homeostasis. The PI3K enzyme family is divided into 3 classes; class I (subdivided into IA and IB), class II (PI3K-C2alpha, PI3K-C2beta and PI3K-C2gamma) and class III PI3K. Expression of these enzymes in human tissue has not been clearly defined. METHODS: In this study, we analysed the immunohistochemical topographical expression profile of class IA (anti-p85 adaptor) and class II PI3K (PI3K-C2alpha and PI3K-C2beta) enzymes in 104 formalin-fixed, paraffin embedded normal adult human (age 33-71 years, median 44 years) tissue specimens including those from the gastrointestinal, genitourinary, hepatobiliary, endocrine, integument and lymphoid systems. Antibody specificity was verified by Western blotting of cell lysates and peptide blocking studies. Immunohistochemistry intensity was scored from undetectable to strong. RESULTS: PI3K enzymes were expressed in selected cell populations of epithelial or mesenchymal origin. Columnar epithelium and transitional epithelia were reactive but mucous secreting and stratified squamous epithelia were not. Mesenchymal elements (smooth muscle and endothelial cells) and glomerular epithelium were only expressed PI3K-C2alpha while ganglion cells expressed p85 and PI3K-C2beta. All three enzymes were detected in macrophages, which served as an internal positive control. None of the three PI3K isozymes was detected in the stem cell/progenitor compartments or in B lymphocyte aggregates. CONCLUSIONS: Taken together, these data suggest that PI3K enzyme distribution is not ubiquitous but expressed selectively in fully differentiated, non-proliferating cells. Identification of the normal in vivo expression pattern of class IA and class II PI3K paves the way for further analyses which will clarify the role played by these enzymes in inflammatory, neoplastic and other human disease conditions.

SELECTION OF CITATIONS
SEARCH DETAIL
...