Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
iScience ; 24(12): 103509, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34934928

ABSTRACT

Although hydroxychloroquine (HCQ) has long been used to treat autoimmune diseases, its mechanism of action remains poorly understood. In CD4 T-cells, we found that a clinically relevant concentration of HCQ inhibited the mitochondrial antioxidant system triggered by TCR crosslinking, leading to increased mitochondrial superoxide, impaired activation-induced autophagic flux, and reduced proliferation of CD4 T-cells. In antigen-presenting cells, HCQ also reduced constitutive activation of the endo-lysosomal protease legumain and toll-like receptor 9, thereby reducing cytokine production, but it had little apparent impact on constitutive antigen processing and peptide presentation. HCQ's effects did not require endo-lysosomal pH change, nor impaired autophagosome-lysosome fusion. We explored the clinical relevance of these findings in patients with celiac disease-a prototypic CD4 T-cell-mediated disease-and found that HCQ limits ex vivo antigen-specific T cell responses. We report a T-cell-intrinsic immunomodulatory effect from HCQ and suggest potential re-purposing of HCQ for celiac disease.

2.
Circulation ; 138(23): 2648-2661, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30571257

ABSTRACT

BACKGROUND: Acute rheumatic fever (ARF) and rheumatic heart disease are autoimmune consequences of group A streptococcus infection and remain major causes of cardiovascular morbidity and mortality around the world. Improved treatment has been stymied by gaps in understanding key steps in the immunopathogenesis of ARF and rheumatic heart disease. This study aimed to identify (1) effector T cell cytokine(s) that might be dysregulated in the autoimmune response of patients with ARF by group A streptococcus, and (2) an immunomodulatory agent that suppresses this response and could be clinically translatable to high-risk patients with ARF. METHODS: The immune response to group A streptococcus was analyzed in peripheral blood mononuclear cells from an Australian Aboriginal ARF cohort by a combination of multiplex cytokine array, flow cytometric analysis, and global gene expression analysis by RNA sequencing. The immunomodulatory drug hydroxychloroquine was tested for effects on this response. RESULTS: We found a dysregulated interleukin-1ß-granulocyte-macrophage colony-stimulating factor (GM-CSF) cytokine axis in ARF peripheral blood mononuclear cells exposed to group A streptococcus in vitro, whereby persistent interleukin-1ß production is coupled to overproduction of GM-CSF and selective expansion of CXCR3+CCR4-CCR6- CD4 T cells. CXCR3+CCR4-CCR6- CD4 T cells are the major source of GM-CSF in human CD4 T cells and CXCL10, a CXCR3 ligand and potent T helper 1 chemoattractant, was elevated in sera from patients with ARF. GM-CSF has recently emerged as a key T cell-derived effector cytokine in numerous autoimmune diseases, including myocarditis, and the production of CXCL10 may explain selective trafficking of these cells to the heart. We provide evidence that interleukin-1ß amplifies the expansion of GM-CSF-expressing CD4 T cells, which is effectively suppressed by hydroxychloroquine. RNA sequencing showed shifts in gene expression profiles and differentially expressed genes in peripheral blood mononuclear cells derived from patients at different clinical stages of ARF. CONCLUSIONS: Given the safety profile of hydroxychloroquine and its clinical pedigree in treating autoimmune diseases such as rheumatoid arthritis, where GM-CSF plays a pivotal role, we propose that hydroxychloroquine could be repurposed to reduce the risk of rheumatic heart disease after ARF.


Subject(s)
Gene Expression Regulation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hydroxychloroquine/pharmacology , Interleukin-1beta/metabolism , Rheumatic Fever/pathology , Adolescent , Adult , C-Reactive Protein/analysis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Child , Cytokines/analysis , Cytokines/genetics , Cytokines/metabolism , Female , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Rheumatic Fever/metabolism , Streptococcus pyogenes/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Young Adult
3.
Nat Commun ; 9(1): 3728, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30214011

ABSTRACT

Anti-microbial signaling pathways are normally triggered by innate immune receptors when detecting pathogenic microbes to provide protective immunity. Here we show that the inflammasome sensor Nlrp1 aggravates DSS-induced experimental mouse colitis by limiting beneficial, butyrate-producing Clostridiales in the gut. The colitis-protective effects of Nlrp1 deficiency are thus reversed by vancomycin treatment, but recapitulated with butyrate supplementation in wild-type mice. Moreover, an activating mutation in Nlrp1a increases IL-18 and IFNγ production, and decreases colonic butyrate to exacerbate colitis. We also show that, in patients with ulcerative colitis, increased NLRP1 in inflamed regions of the colon is associated with increased IFN-γ. In this context, NLRP1, IL-18 or IFN-γ expression negatively correlates with the abundance of Clostridiales in human rectal mucosal biopsies. Our data identify the NLRP1 inflammasome to be a key negative regulator of protective, butyrate-producing commensals, which therefore promotes inflammatory bowel disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Butyrates/metabolism , Clostridiales , Inflammatory Bowel Diseases/metabolism , Interferon-gamma/metabolism , Interleukin-18/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Colitis/metabolism , Colon/pathology , Female , Gastrointestinal Microbiome , Gene Deletion , Humans , Inflammasomes , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NLR Proteins , Rectum/metabolism , Signal Transduction , T-Lymphocytes/cytology , Vancomycin/pharmacology
4.
J Exp Med ; 212(6): 927-38, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26008898

ABSTRACT

Gain-of-function mutations that activate the innate immune system can cause systemic autoinflammatory diseases associated with increased IL-1ß production. This cytokine is activated identically to IL-18 by an intracellular protein complex known as the inflammasome; however, IL-18 has not yet been specifically implicated in the pathogenesis of hereditary autoinflammatory disorders. We have now identified an autoinflammatory disease in mice driven by IL-18, but not IL-1ß, resulting from an inactivating mutation of the actin-depolymerizing cofactor Wdr1. This perturbation of actin polymerization leads to systemic autoinflammation that is reduced when IL-18 is deleted but not when IL-1 signaling is removed. Remarkably, inflammasome activation in mature macrophages is unaltered, but IL-18 production from monocytes is greatly exaggerated, and depletion of monocytes in vivo prevents the disease. Small-molecule inhibition of actin polymerization can remove potential danger signals from the system and prevents monocyte IL-18 production. Finally, we show that the inflammasome sensor of actin dynamics in this system requires caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain, and the innate immune receptor pyrin. Previously, perturbation of actin polymerization by pathogens was shown to activate the pyrin inflammasome, so our data now extend this guard hypothesis to host-regulated actin-dependent processes and autoinflammatory disease.


Subject(s)
Actins/physiology , Cytoskeletal Proteins/metabolism , Hereditary Autoinflammatory Diseases/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Microfilament Proteins/metabolism , Actins/chemistry , Animals , Bone Marrow Cells/cytology , Caspase 1/metabolism , Caspases/metabolism , Clodronic Acid/chemistry , Crosses, Genetic , Culture Media, Conditioned/chemistry , Enzyme-Linked Immunosorbent Assay , Interleukin-18/metabolism , Lipopolysaccharides/metabolism , Liposomes/chemistry , Liver/embryology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Monocytes/cytology , Pyrin , Signal Transduction
5.
Methods Mol Biol ; 1040: 9-18, 2013.
Article in English | MEDLINE | ID: mdl-23852593

ABSTRACT

In addition to several other extracellular substances, phagocytosis of amyloid-forming peptides can perturb cellular homeostasis, leading to activation of the cytoplasmic innate immune receptor NLRP3. Once triggered, NLRP3 forms an inflammasome complex that ultimately cleaves pro-IL-1ß and pro-IL-18 into their mature, secreted forms. Here we describe a protocol by which one type of amyloidogenic peptide, islet amyloid polypeptide (IAPP, otherwise known as amylin) can be prepared and used to stimulate myeloid cells in vitro to engage the NLRP3 inflammasome. Methods for measuring the ensuing inflammasome activation are also described. Although initially soluble, IAPP monomers rapidly aggregate in solution to form oligomers and subsequently insoluble amyloid fibrils. More work is required to examine how this transition influences inflammasome activation for different types of amyloid. The course of amyloid formation and corresponding inflammatory capacity of these pre-fibrillar species following uptake also requires further examination, and we hope that our protocols are useful in these endeavors. While these protocols are restricted to examination of synthetic IAPP, isolation of IAPP aggregates from human and transgenic mouse pancreas will be required to definitively determine the proinflammatory effects of endogenous IAPP oligomers and fibrils.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Islet Amyloid Polypeptide/pharmacology , Animals , Caspase 1/metabolism , Enzyme Activation , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Microscopy, Fluorescence , NLR Family, Pyrin Domain-Containing 3 Protein
6.
PLoS One ; 6(2): e17158, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21364927

ABSTRACT

BACKGROUND: Nuclear factor-κB (NF-κB) is a transcription factor that regulates the transcription of genes involved in a variety of biological processes, including innate and adaptive immunity, stress responses and cell proliferation. Constitutive or excessive NF-κB activity has been associated with inflammatory disorders and higher risk of cancer. In contrast to the mechanisms controlling inducible activation, the regulation of basal NF-κB activation is not well understood. Here we test whether clathrin heavy chain (CHC) contributes to the regulation of basal NF-κB activity in epithelial cells. METHODOLOGY: Using RNA interference to reduce endogenous CHC expression, we found that CHC is required to prevent constitutive activation of NF-κB and gene expression. Immunofluorescence staining showed constitutive nuclear localization of the NF-κB subunit p65 in absence of stimulation after CHC knockdown. Elevated basal p65 nuclear localization is caused by constitutive phosphorylation and degradation of inhibitor of NF-κB alpha (IκBα) through an IκB kinase α (IKKα)-dependent mechanism. The role of CHC in NF-κB signaling is functionally relevant as constitutive expression of the proinflammatory chemokine interleukin-8 (IL-8), whose expression is regulated by NF-κB, was found after CHC knockdown. Disruption of clathrin-mediated endocytosis by chemical inhibition or depletion of the µ2-subunit of the endocytosis adaptor protein AP-2, and knockdown of clathrin light chain a (CHLa), failed to induce constitutive NF-κB activation and IL-8 expression, showing that CHC acts on NF-κB independently of endocytosis and CLCa. CONCLUSIONS: We conclude that CHC functions as a built-in molecular brake that ensures a tight control of basal NF-κB activation and gene expression in unstimulated cells. Furthermore, our data suggest a potential link between a defect in CHC expression and chronic inflammation disorder and cancer.


Subject(s)
Clathrin Heavy Chains/metabolism , Clathrin Heavy Chains/physiology , Endocytosis/physiology , NF-kappa B/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Clathrin Heavy Chains/antagonists & inhibitors , Clathrin Heavy Chains/genetics , Endocytosis/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/physiology , HeLa Cells , Humans , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Interleukin-8/metabolism , Phosphorylation , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured
7.
Immunity ; 33(5): 804-16, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21093316

ABSTRACT

The enteroinvasive bacterium Shigella flexneri uses multiple secreted effector proteins to downregulate interleukin-8 (IL-8) expression in infected epithelial cells. Yet, massive IL-8 secretion is observed in Shigellosis. Here we report a host mechanism of cell-cell communication that circumvents the effector proteins and strongly amplifies IL-8 expression during bacterial infection. By monitoring proinflammatory signals at the single-cell level, we found that the activation of the transcription factor NF-κB and the MAP kinases JNK, ERK, and p38 rapidly propagated from infected to uninfected adjacent cells, leading to IL-8 production by uninfected bystander cells. Bystander IL-8 production was also observed during Listeria monocytogenes and Salmonella typhimurium infection. This response could be triggered by recognition of peptidoglycan and is mediated by gap junctions. Thus, we have identified a mechanism of cell-cell communication that amplifies innate immunity against bacterial infection by rapidly spreading proinflammatory signals via gap junctions to yet uninfected cells.


Subject(s)
Dysentery, Bacillary/immunology , Immunity, Innate , MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Shigella flexneri/immunology , Caco-2 Cells , Cell Communication/immunology , Cell Proliferation , Dysentery, Bacillary/enzymology , Gap Junctions/immunology , Gap Junctions/microbiology , HeLa Cells , Humans , Interleukin-8/analysis , Interleukin-8/immunology , Listeria monocytogenes/immunology , Listeriosis/enzymology , Listeriosis/immunology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Peptidoglycan/immunology , Shigella flexneri/enzymology
8.
PLoS One ; 5(10): e15371, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20976174

ABSTRACT

BACKGROUND: During pathogen infection, innate immunity is initiated via the recognition of microbial products by pattern recognition receptors and the subsequent activation of transcription factors that upregulate proinflammatory genes. By controlling the expression of cytokines, chemokines, anti-bacterial peptides and adhesion molecules, the transcription factor nuclear factor-kappa B (NF-κB) has a central function in this process. In a typical model of NF-κB activation, the recognition of pathogen associated molecules triggers the canonical NF-κB pathway that depends on the phosphorylation of Inhibitor of NF-κB (IκB) by the catalytic subunit IκB kinase ß (IKKß), its degradation and the nuclear translocation of NF-κB dimers. METHODOLOGY: Here, we performed an RNA interference (RNAi) screen on Shigella flexneri-induced NF-κB activation to identify new factors involved in the regulation of NF-κB following infection of epithelial cells by invasive bacteria. By targeting a subset of the human signaling proteome, we found that the catalytic subunit IKKα is also required for complete NF-κB activation during infection. Depletion of IKKα by RNAi strongly reduces the nuclear translocation of NF-κB p65 during S. flexneri infection as well as the expression of the proinflammatory chemokine interleukin-8. Similar to IKKß, IKKα contributes to the phosphorylation of IκBα on serines 32 and 36, and to its degradation. Experiments performed with the synthetic Nod1 ligand L-Ala-D-γ-Glu-meso-diaminopimelic acid confirmed that IKKα is involved in NF-κB activation triggered downstream of Nod1-mediated peptidoglycan recognition. CONCLUSIONS: Taken together, these results demonstrate the unexpected role of IKKα in the canonical NF-κB pathway triggered by peptidoglycan recognition during bacterial infection. In addition, they suggest that IKKα may be an important drug target for the development of treatments that aim at limiting inflammation in bacterial infection.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Peptidoglycan/metabolism , Base Sequence , Blotting, Western , DNA Primers , Enzyme-Linked Immunosorbent Assay , HeLa Cells , Humans , RNA Interference
9.
Phytochemistry ; 71(16): 1832-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20804996

ABSTRACT

Zinc finger-homeodomain proteins (ZF-HDs) have been identified in many plant species. In soybean (Glycine max), GmZF-HD1 functions as a transcription factor that activates the soybean calmodulin isoform-4 (GmCaM-4) gene in response to pathogens. Recently, we reported specific binding of GmZF-HD1 to a 30-nt A/T-rich cis-element which constitutes two repeats of a conserved homeodomain binding site, ATTA, within -1207 to -1128bp of the GmCaM-4 promoter. Herein, homeodomain sequences of the GmZF-HD1 protein were compared to those of other homeodomain proteins and characterized the specificity of DNA sequences in the interaction of the GmCaM-4 promoter with GmZF-HD1 protein. Considering the conservation of homeodomains in plants, the AG sequence within a 30-nt A/T-rich cis-element is required for binding of the GmZF-HD1 protein. Approximately 25-bp of A/T-rich DNA sequences containing an AG sequence is necessary for effective binding to the GmZF-HD1 protein. Taken together, the results support the notion that the GmZF-HD1 protein specifically functions in plant stress signalling by interacting with the promoter of GmCaM-4.


Subject(s)
DNA-Binding Proteins/genetics , Glycine max/genetics , Homeodomain Proteins/genetics , Soybean Proteins/genetics , Transcription Factors/genetics , Zinc Fingers/genetics , Base Sequence , Calmodulin/chemistry , Calmodulin/genetics , Calmodulin/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Protein Isoforms , Soybean Proteins/chemistry , Soybean Proteins/metabolism , Glycine max/chemistry , Glycine max/metabolism , Transcription Factors/metabolism
10.
Mol Cells ; 27(4): 475-80, 2009 Apr 30.
Article in English | MEDLINE | ID: mdl-19390829

ABSTRACT

The transcription of soybean (Glycine max) calmodulin isoform-4 (GmCaM-4) is dramatically induced within 0.5 h of exposure to pathogen or NaCl. Core cis-acting elements that regulate the expression of the GmCaM-4 gene in response to pathogen and salt stress were previously identified, between -1,207 and -1,128 bp, and between -858 and -728 bp, in the GmCaM-4 promoter. Here, we characterized the properties of the DNA-binding complexes that form at the two core cis-acting elements of the GmCaM-4 promoter in pathogen-treated nuclear extracts. We generated GUS reporter constructs harboring various deletions of approximately 1.3-kb GmCaM-4 promoter, and analyzed GUS expression in tobacco plants transformed with these constructs. The GUS expression analysis suggested that the two previously identified core regions are involved in inducing GmCaM-4 expression in the heterologous system. Finally, a transient expression assay of Arabidopsis protoplasts showed that the GmCaM-4 promoter produced greater levels of GUS activity than did the CaMV35S promoter after pathogen or NaCl treatments, suggesting that the GmCaM-4 promoter may be useful in the production of conditional gene expression systems.


Subject(s)
Calmodulin/genetics , Glycine max/genetics , Nicotiana/genetics , Binding Sites , Calmodulin/biosynthesis , DNA-Binding Proteins , Gene Expression Regulation, Plant , Plant Proteins/chemistry , Plant Proteins/genetics , Plants, Genetically Modified , Promoter Regions, Genetic , Protein Isoforms , Sequence Analysis, DNA , Glycine max/metabolism , Stress, Physiological , Nicotiana/metabolism , Transcriptional Activation
11.
PLoS One ; 2(11): e1217, 2007 Nov 21.
Article in English | MEDLINE | ID: mdl-18030348

ABSTRACT

TOR (Target of Rapamycin) is a highly conserved protein kinase and a central controller of cell growth. TOR is found in two functionally and structurally distinct multiprotein complexes termed TOR complex 1 (TORC1) and TOR complex 2 (TORC2). In the present study, we developed a two-dimensional liquid chromatography tandem mass spectrometry (2D LC-MS/MS) based proteomic strategy to identify new mammalian TOR (mTOR) binding proteins. We report the identification of Proline-rich Akt substrate (PRAS40) and the hypothetical protein Q6MZQ0/FLJ14213/CAE45978 as new mTOR binding proteins. PRAS40 binds mTORC1 via Raptor, and is an mTOR phosphorylation substrate. PRAS40 inhibits mTORC1 autophosphorylation and mTORC1 kinase activity toward eIF-4E binding protein (4E-BP) and PRAS40 itself. HeLa cells in which PRAS40 was knocked down were protected against induction of apoptosis by TNFalpha and cycloheximide. Rapamycin failed to mimic the pro-apoptotic effect of PRAS40, suggesting that PRAS40 mediates apoptosis independently of its inhibitory effect on mTORC1. Q6MZQ0 is structurally similar to proline rich protein 5 (PRR5) and was therefore named PRR5-Like (PRR5L). PRR5L binds specifically to mTORC2, via Rictor and/or SIN1. Unlike other mTORC2 members, PRR5L is not required for mTORC2 integrity or kinase activity, but dissociates from mTORC2 upon knock down of tuberous sclerosis complex 1 (TSC1) and TSC2. Hyperactivation of mTOR by TSC1/2 knock down enhanced apoptosis whereas PRR5L knock down reduced apoptosis. PRR5L knock down reduced apoptosis also in mTORC2 deficient cells. The above suggests that mTORC2-dissociated PRR5L may promote apoptosis when mTOR is hyperactive. Thus, PRAS40 and PRR5L are novel mTOR-associated proteins that control the balance between cell growth and cell death.


Subject(s)
Apoptosis/physiology , Carrier Proteins/physiology , Phosphoproteins/physiology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Cell Line , Chromatography, Liquid/methods , Cycloheximide/pharmacology , Humans , Intracellular Signaling Peptides and Proteins , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Proteins , RNA Interference , TOR Serine-Threonine Kinases , Tandem Mass Spectrometry , Transcription Factors/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology
12.
Genome Biol ; 8(7): R142, 2007.
Article in English | MEDLINE | ID: mdl-17640392

ABSTRACT

BACKGROUND: Iron uptake via endocytosis of iron-transferrin-transferrin receptor complexes is a rate-limiting step for cell growth, viability and proliferation in tumor cells as well as non-transformed cells such as activated lymphocytes. Signaling pathways that regulate transferrin uptake have not yet been identified. RESULTS: We surveyed the human signaling proteome for regulators that increase or decrease transferrin uptake by screening 1,804 dicer-generated signaling small interfering RNAs using automated quantitative imaging. In addition to known transport proteins, we identified 11 signaling proteins that included a striking signature set for the phosphatidylinositol-3,4,5-trisphosphate (PtdIns(3,4,5)P3)-target of rapamycin (mTOR) signaling pathway. We show that the PI3K-mTOR signaling pathway is a positive regulator of transferrin uptake that increases the number of transferrin receptors per endocytic vesicle without affecting endocytosis or recycling rates. CONCLUSION: Our study identifies the PtdIns(3,4,5)P3-mTOR signaling pathway as a new regulator of iron-transferrin uptake and serves as a proof-of-concept that targeted RNA interference screens of the signaling proteome provide a powerful and unbiased approach to discover or rank signaling pathways that regulate a particular cell function.


Subject(s)
Phosphatidylinositol Phosphates/physiology , Protein Kinases/physiology , Proteome/physiology , Proteomics/methods , Transferrin/metabolism , Endocytosis/drug effects , Humans , Protein Kinases/genetics , Protein Transport/drug effects , Proteome/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Signal Transduction , TOR Serine-Threonine Kinases , Transfection
13.
Nucleic Acids Res ; 35(11): 3612-23, 2007.
Article in English | MEDLINE | ID: mdl-17485478

ABSTRACT

Calmodulin (CaM) is involved in defense responses in plants. In soybean (Glycine max), transcription of calmodulin isoform 4 (GmCaM4) is rapidly induced within 30 min after pathogen stimulation, but regulation of the GmCaM4 gene in response to pathogen is poorly understood. Here, we used the yeast one-hybrid system to isolate two cDNA clones encoding proteins that bind to a 30-nt A/T-rich sequence in the GmCaM4 promoter, a region that contains two repeats of a conserved homeodomain binding site, ATTA. The two proteins, GmZF-HD1 and GmZF-HD2, belong to the zinc finger homeodomain (ZF-HD) transcription factor family. Domain deletion analysis showed that a homeodomain motif can bind to the 30-nt GmCaM4 promoter sequence, whereas the two zinc finger domains cannot. Critically, the formation of super-shifted complexes by an anti-GmZF-HD1 antibody incubated with nuclear extracts from pathogen-treated cells suggests that the interaction between GmZF-HD1 and two homeodomain binding site repeats is regulated by pathogen stimulation. Finally, a transient expression assay with Arabidopsis protoplasts confirmed that GmZF-HD1 can activate the expression of GmCaM4 by specifically interacting with the two repeats. These results suggest that the GmZF-HD1 and -2 proteins function as ZF-HD transcription factors to activate GmCaM4 gene expression in response to pathogen.


Subject(s)
Calmodulin/genetics , Glycine max/genetics , Homeodomain Proteins/metabolism , Plant Proteins/metabolism , Promoter Regions, Genetic , Amino Acid Sequence , Binding Sites , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Gene Expression Regulation, Plant , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/metabolism , Phylogeny , Plant Proteins/chemistry , Plant Proteins/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Alignment , Glycine max/metabolism , Glycine max/microbiology , Tandem Repeat Sequences , Transcriptional Activation , Zinc Fingers
14.
Science ; 314(5804): 1458-61, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17095657

ABSTRACT

Many signaling, cytoskeletal, and transport proteins have to be localized to the plasma membrane (PM) in order to carry out their function. We surveyed PM-targeting mechanisms by imaging the subcellular localization of 125 fluorescent protein-conjugated Ras, Rab, Arf, and Rho proteins. Out of 48 proteins that were PM-localized, 37 contained clusters of positively charged amino acids. To test whether these polybasic clusters bind negatively charged phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] lipids, we developed a chemical phosphatase activation method to deplete PM PI(4,5)P2. Unexpectedly, proteins with polybasic clusters dissociated from the PM only when both PI(4,5)P2 and phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] were depleted, arguing that both lipid second messengers jointly regulate PM targeting.


Subject(s)
Cell Membrane/metabolism , GTP Phosphohydrolases/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , ADP-Ribosylation Factors/chemistry , ADP-Ribosylation Factors/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , GTP Phosphohydrolases/chemistry , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Mice , Molecular Sequence Data , NIH 3T3 Cells , Second Messenger Systems , Signal Transduction , Static Electricity , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/metabolism , ras Proteins/chemistry , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism
15.
Curr Biol ; 15(13): 1235-41, 2005 Jul 12.
Article in English | MEDLINE | ID: mdl-16005298

ABSTRACT

Ca(2+) signaling in nonexcitable cells is typically initiated by receptor-triggered production of inositol-1,4,5-trisphosphate and the release of Ca(2+) from intracellular stores. An elusive signaling process senses the Ca(2+) store depletion and triggers the opening of plasma membrane Ca(2+) channels. The resulting sustained Ca(2+) signals are required for many physiological responses, such as T cell activation and differentiation. Here, we monitored receptor-triggered Ca(2+) signals in cells transfected with siRNAs against 2,304 human signaling proteins, and we identified two proteins required for Ca(2+)-store-depletion-mediated Ca(2+) influx, STIM1 and STIM2. These proteins have a single transmembrane region with a putative Ca(2+) binding domain in the lumen of the endoplasmic reticulum. Ca(2+) store depletion led to a rapid translocation of STIM1 into puncta that accumulated near the plasma membrane. Introducing a point mutation in the STIM1 Ca(2+) binding domain resulted in prelocalization of the protein in puncta, and this mutant failed to respond to store depletion. Our study suggests that STIM proteins function as Ca(2+) store sensors in the signaling pathway connecting Ca(2+) store depletion to Ca(2+) influx.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Cell Adhesion Molecules , Endoplasmic Reticulum/metabolism , Fluorescence , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Membrane Proteins/genetics , Mutation/genetics , Neoplasm Proteins/genetics , RNA, Small Interfering/genetics , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2
16.
Plant Physiol ; 135(4): 2150-61, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15310827

ABSTRACT

The Ca(2+)-binding protein calmodulin mediates cellular Ca(2+) signals in response to a wide array of stimuli in higher eukaryotes. Plants express numerous CaM isoforms. Transcription of one soybean (Glycine max) CaM isoform, SCaM-4, is dramatically induced within 30 min of pathogen or NaCl stresses. To characterize the cis-acting element(s) of this gene, we isolated an approximately 2-kb promoter sequence of the gene. Deletion analysis of the promoter revealed that a 130-bp region located between nucleotide positions -858 and -728 is required for the stressors to induce expression of SCaM-4. A hexameric DNA sequence within this region, GAAAAA (GT-1 cis-element), was identified as a core cis-acting element for the induction of the SCaM-4 gene. The GT-1 cis-element interacts with an Arabidopsis GT-1-like transcription factor, AtGT-3b, in vitro and in a yeast selection system. Transcription of AtGT-3b is also rapidly induced within 30 min after pathogen and NaCl treatment. These results suggest that an interaction between a GT-1 cis-element and a GT-1-like transcription factor plays a role in pathogen- and salt-induced SCaM-4 gene expression in both soybean and Arabidopsis.


Subject(s)
Calcium-Binding Proteins/genetics , Glycine max/genetics , Promoter Regions, Genetic/genetics , Amino Acid Sequence , Base Sequence , DNA Primers , Gene Deletion , Molecular Sequence Data , Plant Proteins/genetics , Plant Proteins/metabolism , RNA, Plant/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Glycine max/metabolism , Transcription Factors/metabolism
17.
Biochemistry ; 42(40): 11625-33, 2003 Oct 14.
Article in English | MEDLINE | ID: mdl-14529272

ABSTRACT

Hundreds of proteins involved in signaling pathways contain a Ca(2+)-dependent membrane-binding motif called the C2-domain. However, no small C2-domain proteins consisting of a single C2-domain have been reported in animal cells. We have isolated two cDNA clones, OsERG1a and OsERG1b, that encode two small C2-domain proteins of 156 and 159 amino acids, respectively, from a fungal elicitor-treated rice cDNA library. The clones are believed to have originated from a single gene by alternative splicing. Transcript levels of the OsERG1 gene are dramatically elevated by a fungal elicitor prepared from Magnaporthe grisea or by Ca(2+) ions. The OsERG1 protein produced in Escherichia coli binds to phospholipid vesicles in a Ca(2+)-dependent manner and is translocated to the plasma membrane of plant cells by treatment with either a fungal elicitor or a Ca(2+) ionophore. These results suggest that OsERG1 proteins containing a single C2-domain are involved in plant defense signaling systems.


Subject(s)
Magnaporthe/physiology , Magnaporthe/pathogenicity , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Oryza/chemistry , Oryza/microbiology , Plant Proteins/biosynthesis , Plant Proteins/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Calcium Signaling/physiology , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/microbiology , Cloning, Molecular , Cytosol/chemistry , Cytosol/metabolism , Gene Expression Regulation, Plant , Genes, Plant , Genome, Plant , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Molecular Sequence Data , Oryza/genetics , Phospholipids/chemistry , Phospholipids/metabolism , Plant Diseases/microbiology , Plant Proteins/genetics , Plant Proteins/isolation & purification , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...