Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Cell Sci ; 131(5)2018 03 06.
Article in English | MEDLINE | ID: mdl-29378918

ABSTRACT

TMEM55a (also known as PIP4P2) is an enzyme that dephosphorylates the phosphatidylinositol (PtdIns) PtdIns(4,5)P2 to form PtdIns(5)P in vitro However, the in vivo conversion of the polyphosphoinositide into PtdIns(5)P by the phosphatase has not yet been demonstrated, and the role of TMEM55a remains poorly understood. Here, we found that mouse macrophages (Raw264.7) deficient in TMEM55a showed an increased engulfment of large particles without affecting the phagocytosis of Escherichia coli Transfection of a bacterial phosphatase with similar substrate specificity to TMEM55a, namely IpgD, into Raw264.7 cells inhibited the engulfment of IgG-erythrocytes in a manner dependent on its phosphatase activity. In contrast, cells transfected with PIP4K2a, which catalyzes PtdIns(4,5)P2 production from PtdIns(5)P, increased phagocytosis. Fluorescent TMEM55a transfected into Raw264.7 cells was found to mostly localize to the phagosome. The accumulation of PtdIns(4,5)P2, PtdIns(3,4,5)P3 and F-actin on the phagocytic cup was increased in TMEM55a-deficient cells, as monitored by live-cell imaging. Phagosomal PtdIns(5)P was decreased in the knockdown cells, but the augmentation of phagocytosis in these cells was unaffected by the exogenous addition of PtdIns(5)P. Taken together, these results suggest that TMEM55a negatively regulates the phagocytosis of large particles by reducing phagosomal PtdIns(4,5)P2 accumulation during cup formation.


Subject(s)
Phagocytosis/genetics , Phagosomes/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoinositide Phosphatases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Vesicular Transport Proteins/metabolism , Animals , Cell Membrane/metabolism , Macrophages/metabolism , Mice , Phagosomes/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositols/metabolism , Protein Binding , RAW 264.7 Cells
2.
Genes Cells ; 24(5): 366-376, 2019 May.
Article in English | MEDLINE | ID: mdl-30851234

ABSTRACT

Lysophosphatidylinositol-acyltransferase-1 (LPIAT1) specifically catalyzes the transfer of arachidonoyl-CoA to lysophosphoinositides. LPIAT-/- mice have been shown to have severe defects in the brain and liver; however, the exact molecular mechanisms behind these conditions are not well understood. As immune cells have been implicated in liver inflammation based on disfunction of LPIAT1, we generated Raw264.7 macrophages deficient in LPIAT1, using shRNA and CRISPR/Cas9. The amount of C38:4 species in phosphoinositides, especially in PtdInsP2 , was remarkably decreased in these cells. Unlike in wild-type cells, LPIAT1-deficient cells showed prolonged oscillations of intracellular Ca2+ upon UDP stimulation, which is known to activate phospholipase Cß through the Gq-coupled P2Y6 receptor, even in the absence of extracellular Ca2+ . It is speculated that the prolonged Ca2+ response may be relevant to the increased risk of liver inflammation induced by LPIAT1 disfunction.


Subject(s)
Acyltransferases/metabolism , Calcium Signaling , Acyltransferases/genetics , Animals , Mice , RAW 264.7 Cells
3.
Biol Pharm Bull ; 42(6): 923-928, 2019.
Article in English | MEDLINE | ID: mdl-31155588

ABSTRACT

Macrophages endocytose modified low-density lipoproteins (LDL) vigorously via scavenger receptor A (SR-A) to become foam cells. In the present study, we found that Sac1, a member of the Sac family of phosphoinositide phosphatases, increases the protein level of SR-A and upregulates foam cell formation. Mouse macrophages (RAW264.7) were transfected with short hairpin RNAs (shRNAs) against Sac1. Sac1 knockdown decreased cell surface SR-A levels and impaired acetylated LDL-induced foam cell formation. Transfection of Sac1-knockdown cells with shRNA-resistant flag-Sac1 effectively rescued the expression of SR-A. Glycosylation of SR-A was largely attenuated by Sac1 knockdown, but neither mRNA expression nor protein degradation of SR-A were affected. These results suggest that Sac1 maintains SR-A protein levels by modulating SR-A glycosylation.


Subject(s)
Foam Cells/metabolism , Membrane Proteins/metabolism , Phosphoinositide Phosphatases/metabolism , Scavenger Receptors, Class A/metabolism , Animals , Lipoproteins, LDL/metabolism , Membrane Proteins/genetics , Mice , Phosphoinositide Phosphatases/genetics , RAW 264.7 Cells , RNA, Messenger , RNA, Small Interfering , Scavenger Receptors, Class A/genetics
4.
Exp Cell Res ; 357(2): 252-259, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28552585

ABSTRACT

The findings of this study suggest that the phosphoinositide phosphatase Sac3 maintains the protein level of scavenger receptor A (SR-A) and regulates foam cell formation. RAW264.7 macrophages were transfected with short hairpin RNAs that target Sac3. The knockdown decreased the level of the cell surface SR-A and suppressed the acetylated low density lipoprotein-induced foam cell formation. The associated regulator of PIKfyve (ArPIKfyve) is a scaffold protein that protects Sac3 from proteasome-dependent degradation. The knockdown of ArPIKfyve decreased Sac3, cell surface SR-A, and foam cell formation. The knockdown of PIKfyve had no effect on SR-A protein levels. These results suggest that the ArPIKfyve-Sac3 complex regulates SR-A protein levels independently of its effect on PIKfyve activity.


Subject(s)
Flavoproteins/metabolism , Lipid Droplets/metabolism , Macrophages/metabolism , Phosphoinositide Phosphatases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptors, Scavenger/metabolism , Animals , Cell Membrane/metabolism , Flavoproteins/genetics , Gene Knockdown Techniques/methods , Humans , Mice , Phosphoinositide Phosphatases/genetics , Phosphoric Monoester Hydrolases/genetics , RAW 264.7 Cells , Scavenger Receptors, Class A/metabolism
5.
Biochem J ; 464(3): 365-75, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25269936

ABSTRACT

Phosphoinositide 5'-phosphatases have been implicated in the regulation of phagocytosis. However, their precise roles in the phagocytic process are poorly understood. We prepared RAW264.7 macrophages deficient in Inpp5e (shInpp5e) to clarify the role of this lipid phosphatase. In the shInpp5e cells, the uptake of solid particles was increased and the rate of phagosome acidification was accelerated. As expected, levels of PtdIns(3,4,5)P3 and PtdIns(3,4)P2 were increased and decreased respectively, on the forming phagocytic cups of these cells. Unexpectedly, the most prominent consequence of the Inpp5e deficiency was the decreased accumulation of PtdIns3P and Rab5 on the phagosome. The expression of a constitutively active form of Rab5b in the shInpp5e cells rescued the PtdIns3P accumulation. Rab20 has been reported to regulate the activity of Rabex5, a guanine nucleotide exchange factor for Rab5. The association of Rab20 with the phagosome was remarkably abrogated in the shInpp5e cells. Over-expression of Rab20 increased phagosomal PtdIns3P accumulation and delayed its elimination. These results suggest that Inpp5e, through functional interactions with Rab20 on the phagosome, activates Rab5, which, in turn, increases PtdIns3P and delays phagosome acidification.


Subject(s)
Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/physiology , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Acids/metabolism , Animals , Cells, Cultured , Macrophages/metabolism , Mice , Phagocytosis/genetics , Protein Binding , Transfection
6.
J Pharmacol Sci ; 120(4): 270-9, 2012.
Article in English | MEDLINE | ID: mdl-23149576

ABSTRACT

Studies with knockout mice have indicated that the only isoform of phosphoinositide 3-kinase (PI3K) functioning in the oxidative burst of mouse neutrophils in response to heterotrimeric guanine nucleotide-binding protein-coupled receptor (GPCR) agonists is a class-IB PI3K, p110γ. In the present study, we observed that the cells from p110γ(-/-) mice gain a response to N-formyl-Met-Leu-Phe (fMLP) after priming with cytochalasin E. Even the unprimed cells, which show no response to fMLP, produce a significant amount of superoxide, when an effective agonist of the mouse-type fMLP receptors, Trp-Lys-Tyr-Met-Val-D-Met, is used to stimulate the cells. These results suggested that the class-IA isoforms (p110α, p110ß, and p110δ) of PI3K are sufficient to trigger and maintain superoxide production. Examination of the effects of isoform-specific inhibitors suggested that the p110ß isoform is the primary PI3K triggering the response to GPCR agonists when p110γ is absent.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/physiology , Class Ib Phosphatidylinositol 3-Kinase/deficiency , Neutrophils/metabolism , Receptors, G-Protein-Coupled/physiology , Superoxides/metabolism , Animals , Cells, Cultured , Isoenzymes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/agonists
7.
Biochem J ; 423(1): 99-108, 2009 Sep 14.
Article in English | MEDLINE | ID: mdl-19604150

ABSTRACT

PI3K (phosphoinositide 3-kinase) alpha has been implicated in phagocytosis and fluid-phase pinocytosis in macrophages. The subtype-specific role of PI3K in these processes is poorly understood. To elucidate this issue, we made Raw 264.7 cells (a mouse leukaemic monocyte-macrophage cell line) deficient in each of the class-I PI3K catalytic subunits: p110alpha, p110beta, p110delta and p110gamma. Among these cells, only the p110alpha-deficient cells exhibited lower phagocytosis of opsonized and non-opsonized zymosan. The p110alpha-deficient cells also showed the impaired phagocytosis of IgG-opsonized erythrocytes and the impaired fluid-phase pinocytosis of dextran (molecular mass of 40 kDa). Receptor-mediated pinocytosis of DiI (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate)-labelled acetylated low-density lipoprotein and fluid-phase pinocytosis of Lucifer Yellow (molecular mass of 500 Da) were resistant to p110alpha depletion. None of these processes were impaired in cells lacking p110beta, p110delta or p110gamma, but were susceptible to a pan-PI3K inhibitor wortmannin. In cells deficient in the enzymes catalysing PtdIns(3,4,5)P3 breakdown [PTEN (phosphatase and tensin homologue deleted on chromosome 10) or SHIP-1 (Src-homology-2-domain-containing inositol phosphatase-1)], uptake of IgG-opsonized particles was enhanced. These results indicated that phagocytosis and fluid-phase pinocytosis of larger molecules are dependent on the lipid kinase activity of p110alpha, whereas pinocytosis via clathrin-coated and small non-coated vesicles may depend on subtypes of PI3Ks other than class I.


Subject(s)
Macrophages/metabolism , Phagocytosis/genetics , Phosphatidylinositol 3-Kinases/physiology , Pinocytosis/genetics , Animals , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Female , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Zymosan/chemistry , Zymosan/metabolism
8.
Cancer Sci ; 100(8): 1494-501, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19432881

ABSTRACT

Oral administration of hot-water extract of Spirulina, cyanobacterium Spirulina platensis, leads to augmentation of NK cytotoxicity in humans. Here, we applied to syngeneic tumor-implant mice (C57BL/6 versus B16 melanoma) Spirulina to elucidate the mechanism of raising antitumor NK activation. A B16D8 subcell line barely expressed MHC class I but about 50% expressed Rae-1, a ligand for NK activation receptor NKG2D. The Rae-1-positive population of implant B16 melanoma was effectively eliminated in the tumor mass progressed in mice. This antitumor activity was induced in parallel with IFN-gamma and abolished in mice by treatment with asialoGM-1 but not CD8beta Ab, suggesting the effector is NK cell. NK cell activation occurred in the spleen of wild-type mice medicated with Spirulina. This Spirulina-mediated enhanced NK activation was abrogated in MyD88 -/- mice but not in TICAM-1 -/- mice. The NK activating properties of Spirulina depending on MyD88 were confirmed with in vitro bone marrow-derived dendritic cells expressing TLR2/4. In D16D8 tumor challenge studies, the antitumor effect of Spirulina was abolished in MyD88 -/- mice. Hence, orally administered Spirulina enhances tumoricidal NK activation through the MyD88 pathway. Spirulina exerted a synergistic antitumor activity with BCG-cell wall skeleton, which is known to activate the MyD88 pathway via TLR2/4 with no NK enhancing activity. Spirulina and BCG-cell wall skeleton synergistically augmented IFN-gamma production and antitumor potential in the B16D8 versus C57BL/6 system. We infer from these results that NK activation by Spirulina has some advantage in combinational use with BCG-cell wall skeleton for developing adjuvant-based antitumor immunotherapy.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Activation , Melanoma, Experimental/immunology , Spirulina/immunology , Adjuvants, Immunologic/metabolism , Administration, Oral , Animals , BCG Vaccine/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Wall/immunology , Dendritic Cells/immunology , Drug Synergism , Female , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Powders , Specific Pathogen-Free Organisms , Spleen/cytology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
9.
J Biochem ; 166(2): 175-185, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31329883

ABSTRACT

TMEM55B is first identified as phosphatidylinositol-4,5-P24-phosphatases (PtdIns-4,5-P24-phosphatases) that catalyse dephosphorylation of PtdIns-4,5-P2 to PtdIns-5-P. We demonstrate for the first time that TMEM55B is phosphorylated by Erk/MAPK and that this mechanism participates in regulation of lysosomal clustering. Exposure of RAW264.7 macrophages to various stimuli induces phosphorylation of TMEM55B on Ser76 and Ser169, sites corresponding to consensus sequences (PX(S/T)P) for phosphorylation by MAPK. Of these stimuli, Toll-like receptor ligands most strongly induce TMEM55B phosphorylation, and this is blocked by the MEK1/2 inhibitor U0126. However, phosphorylation does not impact intrinsic phosphatase activity of TMEM55B. TMEM55B has recently been implicated in starvation induced lysosomal translocation. Amino acid starvation induces perinuclear lamp1 clustering in RAW264.7 macrophages, which was attenuated by shRNA-mediated knock-down or CRISPR/Cas9-mediated knock-out of TMEM55B. Cells exposed to U0126 also exhibit attenuated lamp1 clustering. Overexpression of TMEM55B but not TMEM55A notably enhances lamp1 clustering, with TMEM55B mutants (lacking phosphorylation sites or mimicking the phosphorylated state) exhibiting lower and higher efficacies (respectively) than wild-type TMEM55B. Collectively, results suggest that phosphorylation of TMEM55B by Erk/MAPK impacts lysosomal dynamics.


Subject(s)
Lysosomes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphoinositide Phosphatases/chemistry , Phosphoinositide Phosphatases/metabolism , Animals , Mice , Phosphorylation , RAW 264.7 Cells
10.
J Biochem ; 165(1): 75-84, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30295876

ABSTRACT

PIKfyve phosphorylates PtdIns(3)P to PtdIns(3, 5)P2. One of the best characterized effector downstream of PtdIns(3, 5)P2 is a lysosomal Ca2+ channel, TRPML1. Although it has been reported that TRPML1 is involved in phagosome-lysosome fusion, the relevance of the Ca2+ channel in phagosome acidification has been denied. In this article, however, we demonstrated that the phagosome acidification was dependent on TRPML1. Based on the classical idea that Fluorescein isothiocyanate (FITC)-fluorescence is highly sensitive to acidic pH, we could estimate the phagosome acidification by time laps imaging. FITC-zymosan fluorescence that was engulfed by macrophages, decreased immediately after the uptake while the extinction of FITC-zymosan fluorescence was delayed in PIKfyve-deficient cells. The acidification arrest was completely rescued in the presence of Ca2+ ionophore A23187. Cells treated with a PIKfyve inhibitor, apilimod, also showed delayed phagosome acidification but were rescued by the overexpression of TRPML1. Additionally, TRPML1 agonist, ML-SA1 was effective to acidify the phagosome in PIKfyve-deficient cells. Another phenotype observed in PIKfyve-deficient cells is vacuole formation. Unexpectedly, enlarged vacuole formation in PIKfyve-deficient cells was not rescued by Ca2+ or over expression of TRPML1. It is likely that the acidification and vacuolation arrest is bifurcating downstream of PIKfyve.


Subject(s)
Acids/metabolism , Calcium Channels/metabolism , Endosomes/metabolism , Phagosomes/metabolism , Phosphatidylinositol 3-Kinases/physiology , Transient Receptor Potential Channels/metabolism , Vacuoles/metabolism , Animals , Calcium/metabolism , Enzyme Inhibitors/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Fluorescence , Hydrogen-Ion Concentration , Ionophores/administration & dosage , Macrolides/pharmacology , Mice , Phosphoinositide-3 Kinase Inhibitors , RAW 264.7 Cells , Time-Lapse Imaging , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors
11.
Biochim Biophys Acta ; 1770(4): 687-93, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17276010

ABSTRACT

Menadione (vitamin K(3)) has been shown to activate Erk in several cell lines. This effect has been shown to be due to the activation of EGF receptors (EGFR) as a result of inhibition of some protein tyrosine phosphatases. In the present study, we examined the effects of menadione on Akt in Chinese hamster ovary cells. The phosphorylation of Akt by menadione was not inhibited by AG1478, an inhibitor of EGFR. Menadione inhibited the lipid phosphatase activity of PTEN in a cell-free system. In an intact cell system, menadione inhibited the effect of transfected PTEN on Akt. Thus, one mechanism of its action was considered the accelerated activation of Akt through inhibition of PTEN. This was not the sole mechanism responsible for the EGFR-independent activation of Akt, because menadione attenuated the rate of Akt dephosphorylation even in PTEN-null PC3 cells. The decelerated inactivation of Akt, probably through inhibition of some tyrosine phosphatases, was considered another mechanism of its action.


Subject(s)
PTEN Phosphohydrolase/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Vitamin K 3/pharmacology , Vitamins/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Time Factors , Transfection , cdc25 Phosphatases/antagonists & inhibitors , cdc25 Phosphatases/metabolism
12.
J Biochem ; 143(3): 295-302, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079163

ABSTRACT

The activation of interleukin 1 receptor-associated kinase (IRAK)-1 is a key event in the transmission of signals from Toll-like receptors (TLRs). The catalytic activity of the protein kinase is not essential for its ability to activate nuclear factor (NF) kappaB, because transfection of a kinase-dead mutant of IRAK-1 (IRAK-1KD) is able to activate NF-kappaB in HEK293T cells. In the present study, we observed that the effect of IRAK-1KD was impaired by simultaneous expression of IRAK-4. The effect of IRAK-4 was accompanied by the phosphorylation and degradation of IRAK-1KD. Expression of IRAK-4KD instead of IRAK-4 did not cause these events. In IRAK-4-deficient Raw264.7 macrophages that were prepared by introducing short-hairpin RNA probes, the basal level of IRAK-1 was increased markedly. Stimulation of these cells with TLR ligands did not cause the degradation of IRAK-1, which was clearly observed in the parent cells. These results suggested that the expression of IRAK-4 alone is sufficient to cause the degradation of IRAK-1; the autophosphorylation of IRAK-1 is not necessary to terminate the TLR-induced activation of NF-kappaB. IRAK-4 has an ability to induce the degradation of IRAK-1 in addition to its role as an activator of IRAK-1.


Subject(s)
Feedback, Physiological , Interleukin-1 Receptor-Associated Kinases/metabolism , NF-kappa B/metabolism , Protein Processing, Post-Translational , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Feedback, Physiological/drug effects , Humans , I-kappa B Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects
13.
Mol Immunol ; 44(9): 2257-64, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17161867

ABSTRACT

Toll-like receptor (TLR) family members recognize specific molecular patterns within pathogens. Signaling through TLRs results in a proximal event that involves direct binding of adaptor proteins to the receptors. We observed that TIRAP/Mal, an adaptor protein for TLR2 and TLR4, binds protein kinase Cdelta (PKCdelta). TIRAP/Mal GST-fusion protein and a TIRAP/Mal antibody were able to precipitate PKCdelta from rat peritoneal macrophage and THP1 cell lysates. Truncation mutants of TIRAP/Mal showed that the TIR domain of TIRAP/Mal is responsible for binding. TLR2- and TLR4-mediated phosphorylation of p38 MAPK, IKK, and IkappaB in RAW264.7 cells were abolished by depletion of PKCdelta. These results suggest that PKCdelta binding to TIRAP/Mal promotes TLR signaling events.


Subject(s)
Membrane Glycoproteins/metabolism , Protein Kinase C-delta/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Animals , Enzyme Activation/drug effects , Female , Glutathione Transferase/metabolism , Humans , I-kappa B Kinase/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/chemistry , Mice , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Protein Binding/drug effects , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary/drug effects , Rats , Receptors, Interleukin-1/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Toll-Like Receptors/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
14.
Cancer Sci ; 98(12): 1936-42, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17888032

ABSTRACT

Interleukin (IL)-12 and IL-18 are secreted by myeloid cells activated with adjuvants such as Bacillus Calmette-Guérin (BCG) cell wall. They induce T-helper 1 polarization in the host immune system and upregulate production of lymphocyte interferon-gamma, which leads to the induction of an antitumor gene program. It has been reported that humans have an immune system that more closely resembles that of the guinea pig in adjuvant-response features rather than the mouse system, which prevents the mouse results being extrapolated to human immunotherapy. Here we have constructed a tumor-implant system in guinea pigs to evaluate the antitumor potential of guinea pig IL-12 (gpIL-12) and guinea pig IL-18 (gpIL-18). Purified recombinant gpIL-12 and gpIL-18 were prepared and applied intraperitoneally to tumor-bearing (line 10 hepatoma) guinea pigs as the basis of the adjuvant immunotherapy. Intraperitoneal administration of gpIL-12 and gpIL-18 led to retardation of primary tumor growth and suppression of lymph-node metastasis in tumor-bearing guinea pigs. The permissible range of IL-12 appeared wider in guinea pigs than in mice. Even at an IL-12 dose higher than that in mice, there was no evidence of side-effects until day 26, when the guinea pigs were killed. gpIL-18 augmented the antitumor effect of gpIL-12 but exerted less ability to suppress lymph-node metastasis. The effects of gpIL-12 and gpIL-18 on the tumors implanted in guinea pigs will encourage us to use IL-12- and IL-18-inducible adjuvants for immunotherapy in human patients with solid cancer.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Interleukin-12/therapeutic use , Interleukin-18/therapeutic use , Liver Neoplasms/drug therapy , Recombinant Proteins/therapeutic use , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Guinea Pigs , Humans , Immunotherapy/methods , Interleukin-12/genetics , Interleukin-18/genetics , Liver Neoplasms/pathology , Toll-Like Receptors/immunology
15.
Innate Immun ; 23(4): 401-409, 2017 05.
Article in English | MEDLINE | ID: mdl-28409542

ABSTRACT

The relative abundance of phosphoinositide (PI) species on the phagosome membrane fluctuates over the course of phagocytosis. PtdIns(3,4,5)P3 and PtdIns(3,4)P2 rapidly increase in the forming of the phagocytic cup, following which they disappear after sealing of the cup. In the present study, we monitored the clearance of these PI species using the enhanced green fluorescent protein-fused pleckstrin homology domain of Akt, a fluorescence probe that binds both PtdIns(3,4,5)P3 and PtdIns(3,4)P2 in Raw 264.7 macrophages. The clearance of PIs was much faster when the phagocytosed particles were coated with IgG. The effect of IgG was not observed in the macrophages deficient in FcγRIIb, an inhibitory IgG receptor. To identify the lipid phosphatases responsible for the FcγRIIb-accelerated PI clearance, we prepared a panel of lipid phosphatase-deficient cells. The lack of a PI 5-phosphatase Src homology 2 domain-containing inositol-5-phosphatase (SHIP)1 or SHIP2 impaired the FcγRIIb-accelerated clearance of PIs. The lack of a PI 4-phosphatase Inpp4a also impaired the accelerated PIs clearance. In the FcγRIIb- and Inpp4a-deficient cells, acidification of the formed phagosome was slowed. These results suggested that FcγRIIb drives the sequential dephosphorylation system comprising SHIPs and Inpp4a, and accelerates phagosome acidification.


Subject(s)
Macrophages/metabolism , Oncogene Protein v-akt/metabolism , Phagocytosis , Phagosomes/metabolism , Phosphoric Monoester Hydrolases/metabolism , Receptors, IgG/metabolism , Animals , Hydrogen-Ion Concentration , Immunoglobulin G/metabolism , Macrophages/immunology , Mice , Oncogene Protein v-akt/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Binding , RAW 264.7 Cells , RNA, Small Interfering/genetics , Receptors, IgG/genetics
16.
Biochem J ; 392(Pt 3): 607-14, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16091017

ABSTRACT

We prepared CHO (Chinese hamster ovary) cells expressing both IR (insulin receptor) and A1R (A1 adenosine receptor). Treatment of the cells with insulin or PIA [N6-(2-phenylisopropyl)adenosine], a specific A(1)R agonist increased Akt activity in the cells in a PI3K- (phosphoinositide 3-kinase) dependent manner. Transfection of p110beta into the cells augmented the action of PIA with little effect on insulin. Introduction of a pH1 vector producing shRNA (short hairpin RNA) that targets p110beta abolished PIA-induced Akt activation. By contrast, an shRNA probe targeting p110alpha did not impair the effects of PIA. The effect of PIA in p110alpha-deficient cells was attenuated effectively by both Deltap85 and betaARK-CT (beta-adrenergic receptor kinase-C-terminal peptide). A Deltap85-derived protein possessing point mutations in its two SH2 domains did not impair PIA action. These results suggest that tyrosine-phosphorylated proteins and Gbetagamma (betagamma subunits of GTP-binding protein) are necessary for the specific function of p110beta in intact cells. The p110beta-middle (middle part of p110beta) may play an important role in signal reception from GPCRs (GTP-binding-protein-coupled receptor), because transfection of the middle part impaired PIA sensitivity.


Subject(s)
GTP-Binding Proteins/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Adenosine A1/metabolism , Animals , CHO Cells , Cattle , Cricetinae , Enzyme Activation , Gene Expression Regulation , Humans , Insulin , Mice , Phosphatidylinositol Phosphates/metabolism , Receptor, Insulin/metabolism
17.
Innate Immun ; 22(6): 444-51, 2016 08.
Article in English | MEDLINE | ID: mdl-27252170

ABSTRACT

Phosphatidylinositol 3-kinase (PI3K)/Akt signaling has been implicated in the anti-inflammatory response in a mouse model of endotoxemia and sepsis. The present study focused on the role of inositol polyphosphate-4-phosphatase type I (Inpp4a), which dephosphorylates PtdIns(3,4)P2 to PtdIns(3)P, in bacterial infections. We prepared myeloid cell-specific Inpp4a-conditional knockout mice. Macrophages from these mice showed increased Akt phosphorylation and reduced production of inflammatory cytokines in response to LPS or Escherichia coli in vitro The Inpp4a knockout mice survived for a shorter time than wild type mice after i.p. infection with E. coli, with less production of inflammatory cytokines. Additionally, E. coli clearance from blood and lung was significantly impaired in the knockout mice. A likely mechanism is that the Inpp4a-catalyzed dephosphorylation of PtdIns(3,4)P2 down-regulates Akt pathways, which, in turn, increases the production of inflammatory mediators. This mechanism at least fits the decreased E. coli clearance and short survival in the Inpp4a knockout mice.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/physiology , Lung/immunology , Macrophages, Peritoneal/physiology , Peritonitis/immunology , Phosphoric Monoester Hydrolases/metabolism , Shock, Septic/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Lung/microbiology , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peritonitis/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Shock, Septic/genetics , Signal Transduction
18.
Mol Immunol ; 38(9): 689-700, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11858824

ABSTRACT

Human CD46, formerly membrane cofactor protein (MCP), binds and inactivates complement C3b and serves as a receptor for measles virus (MV), thereby protecting cells from homologous complement and sustaining systemic viral infection. CD46 on activated macrophages (Mphi) but not intact monocytes is presumed to be the factor responsible for virus-mediated immune modulation including down-regulation of IL-12 production. As CD46 is expressed on both Mphi and monocytes, the molecular mechanisms responsible for these distinct immune responses remain largely unknown. Here, we found that peripheral blood monocytes treated for 5--8 days with GM-CSF (i.e. mature Mphi) acquired the capacity to assemble CD9, alpha3-beta1 integrin and the tyrosine phosphatase SHP-1 with their CD46. Prior to this maturation stage, Mphi expressed sufficient amounts of CD9 and CD46 but showed no such complex formation, and as in intact monocytes MV replication was markedly suppressed. By flow cytometry and confocal microscopy, the complex was found to assemble on the surface in cells treated with approximately 6 days with GM-CSF but not for approximately 2 days. Notably, an alternative MV receptor SLAM CDw150 was neither expressed nor recruited to this complex throughout GM-CSF-mediated Mphi differentiation. These responses and molecular links were not reproduced in the hamster cell line CHO expressing human CD46 although these cells acquired high susceptibility to MV. Based on these observations, MV susceptibility in human myeloid lineages appears not to be as simple as that observed in human CD46-transfected non-myeloid cells. The molecular complex involving CD46 may confer high MV permissiveness leading to immune modulation in Mphi.


Subject(s)
Antigens, CD/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Integrins/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Antigens, CD/immunology , CHO Cells , Cell Differentiation/drug effects , Cricetinae , Female , Flow Cytometry , Humans , Integrin alpha3beta1 , Integrins/immunology , Intracellular Signaling Peptides and Proteins , Macrophages/drug effects , Macrophages/immunology , Membrane Cofactor Protein , Membrane Glycoproteins/immunology , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/immunology , Tetraspanin 29
19.
PLoS One ; 10(11): e0142091, 2015.
Article in English | MEDLINE | ID: mdl-26535897

ABSTRACT

Phagocytosis is a highly conserved process whereby phagocytic cells engulf pathogens and apoptotic bodies. The present study focused on the role of inositol polyphosphate-4-phosphatase type I (Inpp4a) in phagocytosis. Raw264.7 cells that express shRNA against Inpp4a (shInpp4a cells) showed significantly increased phagocytic activity. The introduction of shRNA-resistant human Inpp4a abolished this increase. Macrophages from Inpp4a knockout mice showed similar increases in the phagocytic activity. Inpp4a was recruited to the phagosome membrane by a mechanism other than the direct interaction with Rab5. PtdIns(3,4)P2 increased on the phagosome of shInpp4a cells, while PtdIns(3)P significantly decreased. The results indicate that Inpp4a negatively regulates the phagocytic activity of macrophages as a member of the sequential dephosphorylation system that metabolizes phagosomal PtdIns(3,4,5)P3 to PtdIns(3)P.


Subject(s)
Cell Membrane/metabolism , Macrophages/metabolism , Phagocytosis , Phagosomes/metabolism , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/physiology , Animals , Blotting, Western , Cells, Cultured , Female , Humans , Macrophages/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation
20.
Int Immunopharmacol ; 2(4): 423-34, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11962722

ABSTRACT

Spirulina platensis is a cyanobacterial species that is surmised to potentiate the immune system leading to suppression of cancer development and viral infection. Here, we identified the molecular mechanism of the human immune potentiating capacity of Spirulina by analyzing blood cells of volunteers with pre and post oral administration of hot water extract of Spirulina. NK functions represented by IFN gamma production and cytolysis were enhanced after administration of Spirulina in >50% subjects. IFN gamma was produced in an IL-12/IL-18-dependent fashion. In vitro stimulation of blood cells with BCG cell wall skeleton (CWS) allowed more potent IL-12 p40 production in cells from volunteers given Spirulina than in cells without pre-exposure to Spirulina. As BCG-CWS serves as a ligand for Toll-like receptor (TLR) 2 and 4 to raise the maturation stage of monocytes/macrophages, Spirulina may be involved in the signaling responses through Toll in blood cells even when orally administered. These observations indicated that in humans Spirulina acts directly on myeloid lineages and either directly or indirectly on NK cells. The presence of co-operative IL-12 and IL-18 is critically important for NK-mediated IFN gamma production.


Subject(s)
Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Cytotoxicity, Immunologic , Immunity, Innate/immunology , Interferons/biosynthesis , Killer Cells, Natural/immunology , Administration, Oral , Adult , Cyanobacteria/chemistry , Cyanobacteria/immunology , Flow Cytometry , Humans , Interferon Inducers/administration & dosage , Interferon Inducers/immunology , Interferons/immunology , Interleukin-12/biosynthesis , Interleukin-12/chemistry , Interleukin-12/immunology , Interleukin-18/immunology , Male , Middle Aged , Protein Subunits , Receptors, Interleukin/metabolism , Spirulina
SELECTION OF CITATIONS
SEARCH DETAIL