Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Biochem J ; 479(3): 225-243, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35015072

ABSTRACT

The ability of the cellular immune system to discriminate self from foreign antigens depends on the appropriate calibration of the T cell receptor (TCR) signalling threshold. The lymphocyte homeostatic cytokine interleukin 7 (IL-7) is known to affect TCR thresholding, but the molecular mechanism is not fully elucidated. A better understanding of this process is highly relevant in the context of autoimmune disease therapy and cancer immunotherapy. We sought to characterise the early signalling events attributable to IL-7 priming; in particular, the altered phosphorylation of signal transduction proteins and their molecular localisation to the TCR. By integrating high-resolution proximity- phospho-proteomic and imaging approaches using primary T cells, rather than engineered cell lines or an in vitro expanded T cell population, we uncovered transduction events previously not linked to IL-7. We show that IL-7 leads to dephosphorylation of cytohesin interacting protein (CYTIP) at a hitherto undescribed phosphorylation site (pThr280) and alters the co-localisation of cytohesin-1 with the TCR and LFA-1 integrin. These results show that IL-7, acting via CYTIP and cytohesin-1, may impact TCR activation thresholds by enhancing the co-clustering of TCR and LFA-1 integrin.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Interleukin-7/pharmacology , Lymphocyte Function-Associated Antigen-1/metabolism , Proteome/metabolism , Proteomics/methods , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/drug effects , T-Lymphocytes/immunology , Transcription Factors/metabolism , Actin Cytoskeleton/metabolism , Blood Donors , Cells, Cultured , Humans , Lymphocyte Activation/drug effects , Phosphorylation/drug effects , Recombinant Proteins/pharmacology , Threonine/metabolism
2.
J Immunotoxicol ; 18(1): 61-73, 2021 12.
Article in English | MEDLINE | ID: mdl-33956565

ABSTRACT

There is a large, unmet medical need to treat chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis and other respiratory diseases. New modalities are being developed, including gene therapy which treats the disease at the DNA/RNA level. Despite recent innovations in non-viral gene therapy delivery for chronic respiratory diseases, unwanted or adverse interactions with immune cells, particularly macrophages, can limit drug efficacy. This review will examine the relationship between the design and fabrication of non-viral nucleic acid nanoparticle (NP) delivery systems and their ability to trigger unwanted immunogenic responses in lung tissues. NP formulated with peptides, lipids, synthetic and natural polymers provide a robust means of delivering the genetic cargos to the desired cells. However NP, or their components, may trigger local responses such as cell damage, edema, inflammation, and complement activation. These effects may be acute short-term reactions or chronic long-term effects like fibrosis, increased susceptibility to diseases, autoimmune disorders, and even cancer. This review examines the relationship between physicochemical properties, i.e. shape, charge, hydrophobicity, composition and stiffness, and interactions of NP with pulmonary immune cells. Inhalation is the ideal route of administration for direct delivery but inhaled NP encounter innate immune cells, such as alveolar macrophages (AM) and dendritic cells (DC), that perceive them as harmful foreign material, interfere with gene delivery to target cells, and can induce undesirable side effects. Recommendations for fabrication and formulation of gene therapies to avoid adverse immunological responses are given. These include fine tuning physicochemical properties, functionalization of the surface of NP to actively target diseased pulmonary cells and employing biomimetics to increase immunotolerance.


Subject(s)
Nanoparticles , Administration, Inhalation , Drug Delivery Systems , Lung , Macrophages, Alveolar
3.
J Leukoc Biol ; 105(6): 1183-1194, 2019 06.
Article in English | MEDLINE | ID: mdl-30942918

ABSTRACT

GM-CSF is important in regulating acute, persistent neutrophilic inflammation in certain settings, including lung injury. Ligand binding induces rapid internalization of the GM-CSF receptor (GM-CSFRα) complex, a process essential for signaling. Whereas GM-CSF controls many aspects of neutrophil biology, regulation of GM-CSFRα expression is poorly understood, particularly the role of GM-CSFRα in ligand clearance and whether signaling is sustained despite major down-regulation of GM-CSFRα surface expression. We established a quantitative assay of GM-CSFRα surface expression and used this, together with selective anti-GM-CSFR antibodies, to define GM-CSFRα kinetics in human neutrophils, and in murine blood and alveolar neutrophils in a lung injury model. Despite rapid sustained ligand-induced GM-CSFRα loss from the neutrophil surface, which persisted even following ligand removal, pro-survival effects of GM-CSF required ongoing ligand-receptor interaction. Neutrophils recruited to the lungs following LPS challenge showed initially high mGM-CSFRα expression, which along with mGM-CSFRß declined over 24 hr; this was associated with a transient increase in bronchoalveolar lavage fluid (BALF) mGM-CSF concentration. Treating mice in an LPS challenge model with CAM-3003, an anti-mGM-CSFRα mAb, inhibited inflammatory cell influx into the lung and maintained the level of BALF mGM-CSF. Consistent with neutrophil consumption of GM-CSF, human neutrophils depleted exogenous GM-CSF, independent of protease activity. These data show that loss of membrane GM-CSFRα following GM-CSF exposure does not preclude sustained GM-CSF/GM-CSFRα signaling and that this receptor plays a key role in ligand clearance. Hence neutrophilic activation via GM-CSFR may play an important role in neutrophilic lung inflammation even in the absence of high GM-CSF levels or GM-CSFRα expression.


Subject(s)
Acute Lung Injury/immunology , Gene Expression Regulation/immunology , Neutrophils/immunology , Pulmonary Alveoli/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Adult , Animals , Cell Line, Tumor , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Disease Models, Animal , Female , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/pathology , Pulmonary Alveoli/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Time Factors
4.
Mol Pharmacol ; 95(2): 222-234, 2019 02.
Article in English | MEDLINE | ID: mdl-30459156

ABSTRACT

The transforming growth factor ß (TGFß) superfamily includes TGFß, activins, inhibins, and bone morphogenetic proteins (BMPs). These extracellular ligands have essential roles in normal tissue homeostasis by coordinately regulating cell proliferation, differentiation, and migration. Aberrant signaling of superfamily members, however, is associated with fibrosis as well as tumorigenesis, cancer progression, metastasis, and drug-resistance mechanisms in a variety of cancer subtypes. Given their involvement in human disease, the identification of novel selective inhibitors of TGFß superfamily receptors is an attractive therapeutic approach. Seven mammalian type 1 receptors have been identified that have context-specific roles depending on the ligand and the complex formation with the type 2 receptor. Here, we characterize the biologic effects of two transforming growth factor ß receptor 1 (TGFBR1) kinase inhibitors designed to target TGFß signaling. AZ12601011 [2-(2-pyridinyl)-4-(1H-pyrrolo[3,2-c]pyridin-1-yl)-6,7-dihydro-5H-cyclopenta[d]pyrimidine]; structure previously undisclosed] and AZ12799734 [4-({4-[(2,6-dimethyl-3-pyridinyl)oxy]-2-pyridinyl}amino)benzenesulfonamide] (IC50 = 18 and 47 nM, respectively) were more effective inhibitors of TGFß-induced reporter activity than SB-431542 [4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]benzamide] (IC50 = 84 nM) and LY2157299 [4-[2-(6-methylpyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl]quinoline-6-carboxamide monohydrate]] (galunisertib) (IC50 = 380 nM). AZ12601011 inhibited phosphorylation of SMAD2 via the type 1 receptors activin A receptor type 1B (ALK4), TGFBR1, and activin A receptor type 1C (ALK7). AZ12799734, however, is a pan TGF/BMP inhibitor, inhibiting receptor-mediated phosphorylation of SMAD1 by activin A receptor type 1L, bone morphogenetic protein receptor type 1A, and bone morphogenetic protein receptor type 1B and phosphorylation of SMAD2 by ALK4, TGFBR1, and ALK7. AZ12601011 was highly effective at inhibiting basal and TGFß-induced migration of HaCaT keratinocytes and, furthermore, inhibited tumor growth and metastasis to the lungs in a 4T1 syngeneic orthotopic mammary tumor model. These inhibitors provide new reagents for investigating in vitro and in vivo pathogenic processes and the contribution of TGFß- and BMP-regulated signaling pathways to disease states.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Proliferation/drug effects , DNA-Binding Proteins , Mice , NIH 3T3 Cells , Neoplasm Metastasis/pathology , Phosphorylation/drug effects , Signal Transduction/drug effects , Smad2 Protein/metabolism
5.
Respir Res ; 18(1): 159, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28830544

ABSTRACT

BACKGROUND: Increased interferon gamma (IFNγ) release occurs in Chronic Obstructive Pulmonary Disease (COPD) lungs. IFNγ supports optimal viral clearance, but if dysregulated could increase lung tissue destruction. METHODS: The present study investigates which mediators most closely correlate with IFNγ in sputum in stable and exacerbating disease, and seeks to shed light on the spatial requirements for innate production of IFNγ, as reported in mouse lymph nodes, to observe whether such microenvironmental cellular organisation is relevant to IFNγ production in COPD lung. RESULTS: We show tertiary follicle formation in severe disease alters the dominant mechanistic drivers of IFNγ production, because cells producing interleukin-18, a key regulator of IFNγ, are highly associated with such structures. Interleukin-1 family cytokines correlated with IFNγ in COPD sputum. We observed that the primary source of IL-18 in COPD lungs was myeloid cells within lymphoid aggregates and IL-18 was increased in severe disease. IL-18 released from infected epithelium or from activated myeloid cells, was more dominant in driving IFNγ when IL-18-producing and responder cells were in close proximity. CONCLUSIONS: Unlike tight regulation to control infection spread in lymphoid organs, this local interface between IL-18-expressing and responder cell is increasingly supported in lung as disease progresses, increasing its potential to increase tissue damage via IFNγ.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-18/biosynthesis , Lung/metabolism , Lymphocytes/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Severity of Illness Index , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Longitudinal Studies , Lung/pathology , Lymphocytes/pathology , Prospective Studies , Pulmonary Disease, Chronic Obstructive/pathology , Sputum/metabolism
6.
Oncotarget ; 7(50): 81995-82012, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27835901

ABSTRACT

Recent data implicate elevated transforming growth factor-ß (TGFß) signalling in BRAF inhibitor drug-resistance mechanisms, but the potential for targeting TGFß signalling in cases of advanced melanoma has not been investigated. We show that mutant BRAFV600E confers an intrinsic dependence on TGFß/TGFß receptor 1 (TGFBR1) signalling for clonogenicity of murine melanocytes. Pharmacological inhibition of the TGFBR1 blocked the clonogenicity of human mutant BRAF melanoma cells through SMAD4-independent inhibition of mitosis, and also inhibited metastasis in xenografted zebrafish. When investigating the therapeutic potential of combining inhibitors of mutant BRAF and TGFBR1, we noted that unexpectedly, low-dose PLX-4720 (a vemurafenib analogue) promoted proliferation of drug-naïve melanoma cells. Pharmacological or pharmacogenetic inhibition of TGFBR1 blocked growth promotion and phosphorylation of SRC, which is frequently associated with vemurafenib-resistance mechanisms. Importantly, vemurafenib-resistant patient derived cells retained sensitivity to TGFBR1 inhibition, suggesting that TGFBR1 could be targeted therapeutically to combat the development of vemurafenib drug-resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Dioxoles/pharmacology , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacology , Melanoma/drug therapy , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Animals, Genetically Modified , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/genetics , Humans , Melanocytes/drug effects , Melanocytes/enzymology , Melanocytes/pathology , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice, Nude , Mitosis/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Smad4 Protein/genetics , Smad4 Protein/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/pharmacology , Vemurafenib , Xenograft Model Antitumor Assays , Zebrafish
7.
Adv Biol Regul ; 60: 36-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26596865

ABSTRACT

Class I phosphoinositide 3-kinases (PI3Ks) are important regulators of neutrophil migration in response to a range of chemoattractants. Their primary lipid products PtdIns(3,4,5)P3 and PtdIns(3,4)P2 preferentially accumulate near to the leading edge of migrating cells and are thought to act as an important cue organizing molecular and morphological polarization. We have investigated the distribution and accumulation of these lipids independently in mouse neutrophils using eGFP-PH reportersand electron microscopy (EM). We found that authentic mouse neutrophils rapidly polarized their Class I PI3K signalling, as read-out by eGFP-PH reporters, both at the up-gradient leading edge in response to local stimulation with fMLP as well as spontaneously and randomly in response to uniform stimulation. EM studies revealed these events occurred at the plasma membrane, were dominated by accumulation of PtdIns(3,4,5)P3, but not PtdIns(3,4)P2, and were dependent on PI3Kγ and its upstream activation by both Ras and Gßγs.


Subject(s)
Class Ib Phosphatidylinositol 3-Kinase/metabolism , Neutrophils/enzymology , Phosphatidylinositol Phosphates/metabolism , Animals , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , Mice , Neutrophils/metabolism , Protein Transport
8.
PLoS One ; 8(10): e75045, 2013.
Article in English | MEDLINE | ID: mdl-24124465

ABSTRACT

We have addressed the differential roles of class I Phosphoinositide 3-kinases (PI3K) in human breast-derived MCF10a (and iso-genetic derivatives) and MDA-MB 231 and 468 cells. Class I PI3Ks are heterodimers of p110 catalytic (α, ß, δ and γ) and p50-101 regulatory subunits and make the signaling lipid, phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3) that can activate effectors, eg protein kinase B (PKB), and responses, eg migration. The PtdIns(3,4,5)P3-3-phosphatase and tumour-suppressor, PTEN inhibits this pathway. p110α, but not other p110s, has a number of onco-mutant variants that are commonly found in cancers. mRNA-seq data shows that MCF10a cells express p110ß>>α>δ with undetectable p110γ. Despite this, EGF-stimulated phosphorylation of PKB depended upon p110α-, but not ß- or δ- activity. EGF-stimulated chemokinesis, but not chemotaxis, was also dependent upon p110α, but not ß- or δ- activity. In the presence of single, endogenous alleles of onco-mutant p110α (H1047R or E545K), basal, but not EGF-stimulated, phosphorylation of PKB was increased and the effect of EGF was fully reversed by p110α inhibitors. Cells expressing either onco-mutant displayed higher basal motility and EGF-stimulated chemokinesis.This latter effect was, however, only partially-sensitive to PI3K inhibitors. In PTEN(-/-) cells, basal and EGF-stimulated phosphorylation of PKB was substantially increased, but the p110-dependency was variable between cell types. In MDA-MB 468s phosphorylation of PKB was significantly dependent on p110ß, but not α- or δ- activity; in PTEN(-/-) MCF10a it remained, like the parental cells, p110α-dependent. Surprisingly, loss of PTEN suppressed basal motility and EGF-stimulated chemokinesis. These results indicate that; p110α is required for EGF signaling to PKB and chemokinesis, but not chemotaxis; onco-mutant alleles of p110α augment signaling in the absence of EGF and may increase motility, in part, via acutely modulating PI3K-activity-independent mechanisms. Finally, we demonstrate that there is not a universal mechanism that up-regulates p110ß function in the absence of PTEN.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Cell Line , Cell Line, Tumor , Epidermal Growth Factor/pharmacology , Female , Humans , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
9.
PLoS One ; 6(6): e21205, 2011.
Article in English | MEDLINE | ID: mdl-21698257

ABSTRACT

Short chain fatty acids (SCFAs) have recently attracted attention as potential mediators of the effects of gut microbiota on intestinal inflammation. Some of these effects have been suggested to occur through the direct actions of SCFAs on the GPR43 receptor in neutrophils, though the precise role of this receptor in neutrophil activation is still unclear. We show that mouse bone marrow derived neutrophils (BMNs) can chemotax effectively through polycarbonate filters towards a source of acetate, propionate or butyrate. Moreover, we show that BMNs move with good speed and directionality towards a source of propionate in an EZ-Taxiscan chamber coated with fibrinogen. These effects of SCFAs were mimicked by low concentrations of the synthetic GPR43 agonist phenylacetamide-1 and were abolished in GPR43(-/-) BMNs. SCFAs and phenylacetamide-1 also elicited GPR43-dependent activation of PKB, p38 and ERK and these responses were sensitive to pertussis toxin, indicating a role for Gi proteins. Phenylacetamide-1 also elicited rapid and transient activation of Rac1/2 GTPases and phosphorylation of ribosomal protein S6. Genetic and pharmacological intervention identified important roles for PI3Kγ, Rac2, p38 and ERK, but not mTOR, in GPR43-dependent chemotaxis. These results identify GPR43 as a bona fide chemotactic receptor for neutrophils in vitro and start to define important elements in its signal transduction pathways.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Fatty Acids/pharmacology , Neutrophils/drug effects , Receptors, G-Protein-Coupled/physiology , Animals , Blotting, Western , Mice , Mice, Knockout , Neutrophils/cytology , Receptors, G-Protein-Coupled/genetics
10.
Nat Cell Biol ; 9(1): 86-91, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17173040

ABSTRACT

The directional movement of cells in a gradient of external stimulus is termed chemotaxis and is important in many aspects of development and differentiated cell function. Phophoinositide 3-kinases (PI(3)Ks) are thought to have critical roles within the gradient-sensing machinery of a variety of highly motile cells, such as mammalian phagocytes, allowing these cells to respond quickly and efficiently to shallow gradients of soluble stimuli. Our analysis of mammalian neutrophil migration towards ligands such as fMLP shows that, although PtdIns(3,4)P(2) and PtdIns(3,4,5)P(3) accumulate in a PI(3)Kgamma-dependent fashion at the up-gradient leading-edge, this signal is not required for efficient gradient-sensing and gradient-biased movement. PI(3)Kgamma activity is however, a critical determinant of the proportion of cells that can move, that is, respond chemokinetically, in reaction to fMLP. Furthermore, this dependence of chemokinesis on PI(3)Kgamma activity is context dependent, both with respect to the state of priming of the neutrophils and the type of surface on which they are migrating. We propose this effect of PI(3)Kgamma is through roles in the regulation of some aspects of neutrophil polarization that are relevant to movement, such as integrin-based adhesion and the accumulation of polymerized (F)-actin at the leading-edge.


Subject(s)
Neutrophils/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphatidylinositol Phosphates/metabolism , Actins/chemistry , Animals , Cell Movement , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase , Dose-Response Relationship, Drug , Humans , Integrins/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism
11.
J Lipid Res ; 48(3): 726-32, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17130283

ABSTRACT

We describe a novel approach to the relative quantification of phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P(3)] and its application to measure, in neutrophils, the activation of phosphoinositide 3-kinase (PI3K). This protein-lipid overlay-based assay allowed us to confirm and extend the observations, first, that N-formyl-methionyl-leucyl-phenylalanine (fMLP) stimulation of primed human neutrophils leads to a transient and biphasic increase in PtdIns(3,4,5)P(3) levels and, second, that the ability of fMLP to stimulate PtdIns(3,4,5)P(3) accumulation in neutrophils isolated from mice carrying a Ras-insensitive ('DASAA') knock-in of PI3Kgamma (p110gamma(DASAA/DASAA)) is substantially dependent on the Ras binding domain of PI3Kgamma.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Animals , Binding Sites , Cell Line , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase , Humans , Immunoblotting , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Knockout , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Spodoptera
12.
Nat Cell Biol ; 8(11): 1303-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17041586

ABSTRACT

Through their ability to regulate production of the key lipid messenger PtdIns(3,4,5)P(3), the class I phosphatidylinositol-3-OH kinases (PI(3)Ks) support many critical cell responses. They, in turn, can be regulated by cell-surface receptors through signals acting on either their adaptor subunits (for example, through phosphotyrosine or Gbetagammas) or their catalytic subunits (for example, through GTP-Ras). The relative significance of these controlling inputs is undefined in vivo. Here, we have studied the roles of Gbetagammas, the adaptor p101, Ras and the Ras binding domain (RBD) in the control of the class I PI(3)K, PI(3)Kgamma, in mouse neutrophils. Loss of p101 leads to major reductions in the accumulation of PtdIns(3,4,5)P(3), activation of protein kinase B (PKB) and in migration towards G-protein activating ligands in vitro, and to an aseptically inflamed peritoneum in vivo. Loss of sensitivity of PI(3)Kgamma to Ras unexpectedly caused similar reductions, but additionally caused a substantial loss in production of reactive oxygen species (ROS). We conclude that Gbetagammas, p101 and the Ras-RBD interaction all have important roles in the regulation of PI(3)Kgamma in vivo and that they can simultaneously, but differentially, control distinct PI(3)Kgamma effectors.


Subject(s)
GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , ras Proteins/metabolism , Animals , Binding Sites , Blotting, Western/methods , Cell Movement/drug effects , Class I Phosphatidylinositol 3-Kinases , Complement C5a/pharmacology , Dose-Response Relationship, Drug , Female , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/cytology , Neutrophils/drug effects , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology
13.
J Exp Med ; 203(8): 1927-37, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16880254

ABSTRACT

The generation of reactive oxygen species (ROS) by the reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex plays a critical role in the antimicrobial functions of the phagocytic cells of the immune system. The catalytic core of this oxidase consists of a complex between gp91(phox), p22(phox), p47(phox), p67(phox), p40(phox), and rac-2. Mutations in each of the phox components, except p40(phox), have been described in cases of chronic granulomatous disease (CGD), defining their essential role in oxidase function. We sought to establish the role of p40(phox) by investigating the NADPH oxidase responses of neutrophils isolated from p40(phox-/-) mice. In the absence of p40(phox), the expression of p67(phox) is reduced by approximately 55% and oxidase responses to tumor necrosis factor alpha/fibrinogen, immunoglobulin G latex beads, Staphylococcus aureus, formyl-methionyl-leucyl-phenylalanine, and zymosan were reduced by approximately 97, 85, 84, 75, and 30%, respectively. The defect in ROS production by p40(phox-/-) neutrophils in response to S. aureus translated into a severe, CGD-like defect in the killing of this organism both in vitro and in vivo, defining p40(phox) as an essential component in bacterial killing.


Subject(s)
Gene Expression Regulation, Enzymologic , NADPH Oxidases/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Oxidants/metabolism , Phosphoproteins/deficiency , Staphylococcus aureus/immunology , Animals , Bacterial Infections/immunology , Bacterial Infections/microbiology , Blood Cell Count , Cell Adhesion/drug effects , Cell Differentiation , Fibrinogen/metabolism , Mice , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , NADPH Oxidases/genetics , Neutrophils/drug effects , Neutrophils/enzymology , Oxidants/pharmacology , Phagocytosis/drug effects , Phosphoproteins/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Solubility , Tumor Necrosis Factor-alpha/pharmacology
14.
J Immunol ; 176(9): 5314-20, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16621998

ABSTRACT

RhoG is a Rho family small GTPase implicated in cytoskeletal regulation, acting either upstream of or in parallel to Rac1. The precise function(s) of RhoG in vivo has not yet been defined. We have identified a novel role for RhoG in signaling the neutrophil respiratory burst stimulated by G protein-coupled receptor agonists. Bone marrow-derived neutrophils from RhoG knockout (RhoG(-/-)) mice exhibited a marked impairment of oxidant generation in response to C5a or fMLP, but normal responses to PMA or opsonized zymosan and normal bacterial killing. Activation of Rac1 and Rac2 by fMLP was diminished in RhoG(-/-) neutrophils only at very early (5 s) time points (by 25 and 32%, respectively), whereas chemotaxis in response to soluble agonists was unaffected by lack of RhoG. Additionally, fMLP-stimulated phosphorylation of protein kinase B and p38MAPK, activation of phospholipase D, and calcium fluxes were equivalent in wild-type and RhoG(-/-) neutrophils. Our results define RhoG as a critical component of G protein-coupled receptor-stimulated signaling cascades in murine neutrophils, acting either via a subset of total cellular Rac relevant to oxidase activation and/or by a novel and as yet undefined interaction with the neutrophil NADPH oxidase.


Subject(s)
NADPH Oxidases/metabolism , Neutrophils/enzymology , rho GTP-Binding Proteins/metabolism , Animals , Calcium/metabolism , Catecholamines/pharmacology , Cells, Cultured , Chemotaxis, Leukocyte , Enzyme Activation , Gene Expression Regulation, Enzymologic , Humans , Imidazolines/pharmacology , Mice , Mice, Knockout , Neutrophils/cytology , Neutrophils/drug effects , Phospholipase D/metabolism , Signal Transduction , Solubility , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/agonists , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics
15.
Curr Biol ; 15(20): 1867-73, 2005 Oct 25.
Article in English | MEDLINE | ID: mdl-16243035

ABSTRACT

Rac GTPases regulate cytoskeletal structure, gene expression, and reactive oxygen species (ROS) production. Rac2-deficient neutrophils cannot chemotax, produce ROS, or degranulate upon G protein-coupled receptor (GPCR) activation. Deficiency in PI3Kgamma, an upstream regulator of Rac, causes a similar phenotype. P-Rex1, a guanine-nucleotide exchange factor (GEF) for Rac, is believed to link GPCRs and PI3Kgamma to Rac-dependent neutrophil responses. We have investigated the functional importance of P-Rex1 by generating a P-Rex1(-/-) mouse. P-Rex1(-/-) mice are viable and healthy, with apparently normal leukocyte development, but with mild neutrophilia. In neutrophils from P-Rex1(-/-) mice, GPCR-dependent Rac2 activation is impaired, whereas Rac1 activation is less compromised. GPCR-dependent ROS formation is absent in lipopolysaccharide (LPS)-primed P-Rex1(-/-) neutrophils, but less affected in unprimed or TNFalpha-primed cells. Recruitment of P-Rex1(-/-) neutrophils to inflammatory sites is impaired. Surprisingly, chemotaxis of isolated neutrophils is only slightly reduced, with a mild defect in cell speed, but normal polarization and directionality. Secretion of azurophil granules is unaffected. In conclusion, P-Rex1 is an important regulator of neutrophil function by mediating a subset of Rac-dependent neutrophil responses. However, P-Rex1 is not an essential regulator of neutrophil chemotaxis and degranulation.


Subject(s)
Cell Degranulation/physiology , Chemotaxis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Neutrophils/metabolism , Neutrophils/physiology , Actins/metabolism , Animals , Cloning, Molecular , Enzyme Activation/physiology , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Knockout , Neuropeptides/metabolism , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , RAC2 GTP-Binding Protein
16.
Curr Biol ; 15(6): 566-70, 2005 Mar 29.
Article in English | MEDLINE | ID: mdl-15797027

ABSTRACT

A variety of genetic and inhibitor studies have shown that phosphoinositide 3-kinase gamma (PI3Kgamma) plays an essential role in a number of physiological responses, including neutrophil chemotaxis, mast cell degranulation, and cardiac function []. PI3Kgamma is currently thought to be composed of a p110gamma catalytic subunit and a single regulatory subunit, p101. The binding of p110gamma to p101 dramatically increases the activation of the complex by Gbetagamma subunits and, hence, is thought to be critical for the coupling of PI3Kgamma to G protein coupled receptors []. Here, we characterize a new regulatory subunit for PI3Kgamma. p84 is present in human, mouse, chicken, frog, and fugu genomes and is located beside the p101 locus. It is broadly expressed in cells of the murine immune system. Both recombinant and endogenous p84 bind p110gamma specifically and with high affinity. Binding of p84 to p110gamma substantially increases the ability of Gbetagamma to stimulate phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P(3)) production both in vitro and in vivo. However, the p84/p110gamma heterodimer is approximately 4-fold less sensitive to Gbetagammas than p101/p110gamma. Endogenous murine p84 expression is substantially reduced in the absence of p110gamma expression. We conclude that p110gamma has two potential regulatory subunits in vivo, p84 and p101.


Subject(s)
Gene Expression , Phosphatidylinositol 3-Kinases/genetics , Phylogeny , Protein Subunits/genetics , Vertebrates/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Cluster Analysis , DNA, Complementary/genetics , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Humans , Mice , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Conformation , Protein Subunits/metabolism , Sequence Alignment , Sequence Analysis, DNA
17.
Cell ; 108(6): 809-21, 2002 Mar 22.
Article in English | MEDLINE | ID: mdl-11955434

ABSTRACT

Rac, a member of the Rho family of monomeric GTPases, is an integrator of intracellular signaling in a wide range of cellular processes. We have purified a PtdIns(3,4,5)P3-sensitive activator of Rac from neutrophil cytosol. It is an abundant, 185 kDa guanine-nucleotide exchange factor (GEF), which we cloned and named P-Rex1. The recombinant enzyme has Rac-GEF activity that is directly, substantially, and synergistically activated by PtdIns(3,4,5)P3 and Gbetagammas both in vitro and in vivo. P-Rex1 antisense oligonucleotides reduced endogenous P-Rex1 expression and C5a-stimulated reactive oxygen species formation in a neutrophil-like cell line. P-Rex1 appears to be a coincidence detector in PtdIns(3,4,5)P3 and Gbetagamma signaling pathways that is particularly adapted to function downstream of heterotrimeric G proteins in neutrophils.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Phosphatidylinositol Phosphates/metabolism , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Fractionation , Cell Line , Cloning, Molecular , Cytosol/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , In Vitro Techniques , Molecular Sequence Data , Neutrophils/cytology , Neutrophils/metabolism , Phenotype , Phosphoric Monoester Hydrolases/metabolism , Platelet-Derived Growth Factor/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...