Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Cell Sci ; 137(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38264908

ABSTRACT

Activator of G-protein signaling 3 (AGS3; also known as GPSM1), a receptor-independent activator of G-protein signaling, oscillates among defined subcellular compartments and biomolecular condensates (BMCs) in a regulated manner that is likely related to the functional diversity of the protein. We determined the influence of cell stress on the cellular distribution of AGS3 and core material properties of AGS3 BMCs. Cellular stress (oxidative, pHi and thermal) induced the formation of AGS3 BMCs in HeLa and COS-7 cells, as determined by fluorescent microscopy. Oxidative stress-induced AGS3 BMCs were distinct from G3BP1 stress granules and from RNA processing BMCs defined by the P-body protein Dcp1a. Immunoblots indicated that cellular stress shifted AGS3, but not the stress granule protein G3BP1 to a membrane pellet fraction following cell lysis. The stress-induced generation of AGS3 BMCs was reduced by co-expression of the signaling protein Gαi3, but not the AGS3-binding partner DVL2. Fluorescent recovery following photobleaching of individual AGS3 BMCs indicated that there are distinct diffusion kinetics and restricted fluidity for AGS3 BMCs. These data suggest that AGS3 BMCs represent a distinct class of stress granules that serve as a previously unrecognized signal processing node.


Subject(s)
Biomolecular Condensates , Carrier Proteins , Carrier Proteins/metabolism , DNA Helicases , GTP-Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/metabolism , RNA Recognition Motif Proteins , Humans , Animals
2.
J Cell Sci ; 133(17)2020 09 04.
Article in English | MEDLINE | ID: mdl-32737219

ABSTRACT

Activator of G-protein signaling 3 (AGS3, encoded by GPSM1) was discovered as a one of several receptor-independent activators of G-protein signaling, which are postulated to provide a platform for divergence between canonical and noncanonical G-protein signaling pathways. Similarly, Dishevelled (DVL) proteins serve as a point of divergence for ß-catenin-dependent and -independent signaling pathways involving the family of Frizzled (FZD) ligands and cell-surface WNT receptors. We recently discovered the apparent regulated localization of dishevelled-2 (DVL2) and AGS3 to distinct cellular puncta, suggesting that the two proteins interact as part of various cell signaling systems. To address this hypothesis, we asked the following questions: (1) do AGS3 signaling pathways influence the activation of ß-catenin (CTNNB1)-regulated transcription through the WNT-Frizzled-Dishevelled axis, and (2) is the AGS3 and DVL2 interaction regulated? The interaction of AGS3 and DVL2 was regulated by protein phosphorylation, subcellular distribution, and a cell-surface G-protein-coupled receptor. These data, and the commonality of functional system impacts observed for AGS3 and DVL2, suggest that the AGS3-DVL2 complex presents an unexpected path for functional integration within the cell.This article has an associated First Person interview with the first author of the paper.


Subject(s)
GTP-Binding Proteins , Signal Transduction , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Wnt Signaling Pathway
3.
J Immunol ; 202(5): 1510-1520, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30683698

ABSTRACT

Macrophages exist as innate immune subsets that exhibit phenotypic heterogeneity and functional plasticity. Their phenotypes are dictated by inputs from the tissue microenvironment. G-protein-coupled receptors are essential in transducing signals from the microenvironment, and heterotrimeric Gα signaling links these receptors to downstream effectors. Several Gαi-coupled G-protein-coupled receptors have been implicated in macrophage polarization. In this study, we use genetically modified mice to investigate the role of Gαi2 on inflammasome activity and macrophage polarization. We report that Gαi2 in murine bone marrow-derived macrophages (BMDMs) regulates IL-1ß release after activation of the NLRP3, AIM2, and NLRC4 inflammasomes. We show this regulation stems from the biased polarity of Gαi2 deficient (Gnai2 -/-) and RGS-insensitive Gαi2 (Gnai2 G184S/G184S) BMDMs. We determined that although Gnai2 G184S/G184S BMDMs (excess Gαi2 signaling) have a tendency toward classically activated proinflammatory (M1) phenotype, Gnai2-/- BMDMs (Gαi2 deficient) are biased toward alternatively activated anti-inflammatory (M2) phenotype. Finally, we find that Gαi2-deficient macrophages have increased Akt activation and IFN-ß production but defects in ERK1/2 and STAT3 activation after LPS stimulation. Gαi2-deficient macrophages also exhibit increased STAT6 activation after IL-4 stimulation. In summary, our data indicates that excess Gαi2 signaling promotes an M1 macrophage phenotype, whereas Gαi2 signaling deficiency promotes an M2 phenotype. Understanding Gαi2-mediated effects on macrophage polarization may bring to light insights regarding disease pathogenesis and the reprogramming of macrophages for the development of novel therapeutics.


Subject(s)
Cytokines/biosynthesis , GTP-Binding Protein alpha Subunit, Gi2/immunology , Inflammasomes/immunology , Macrophages/immunology , Signal Transduction/immunology , Animals , Cells, Cultured , GTP-Binding Protein alpha Subunit, Gi2/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
4.
J Cell Sci ; 131(23)2018 12 05.
Article in English | MEDLINE | ID: mdl-30404823

ABSTRACT

Activator of G-protein signaling 3 (AGS3, also known as GPSM1) exhibits broad functional diversity and oscillates among different subcellular compartments in a regulated manner. AGS3 consists of a tetratricopeptide repeat (TPR) domain and a G-protein regulatory (GPR) domain. Here, we tested the hypothesis that phosphorylation of the AGS3 GPR domain regulates its subcellular distribution and functionality. In contrast to the cortical and/or diffuse non-homogeneous distribution of wild-type (WT) AGS3, an AGS3 construct lacking all 24 potential phosphorylation sites in the GPR domain localized to cytosolic puncta. This change in localization was revealed to be dependent upon phosphorylation of a single threonine amino acid (T602). The punctate distribution of AGS3-T602A was rescued by co-expression of Gαi and Gαo but not Gαs or Gαq Following treatment with alkaline phosphatase, both AGS3-T602A and WT AGS3 exhibited a gel shift in SDS-PAGE as compared to untreated WT AGS3, consistent with a loss of protein phosphorylation. The punctate distribution of AGS3-T602A was lost in an AGS3-A602T conversion mutant, but was still present upon T602 mutation to glutamate or aspartate. These results implicate dynamic phosphorylation as a discrete mechanism to regulate the subcellular distribution of AGS3 and associated functionality.


Subject(s)
GTP-Binding Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Animals , Phosphorylation , Signal Transduction
5.
J Pharmacol Exp Ther ; 360(3): 424-433, 2017 03.
Article in English | MEDLINE | ID: mdl-28062526

ABSTRACT

Activator of G-protein signaling 4 (AGS4)/G-protein signaling modulator 3 (Gpsm3) contains three G-protein regulatory (GPR) motifs, each of which can bind Gαi-GDP free of Gßγ We previously demonstrated that the AGS4-Gαi interaction is regulated by seven transmembrane-spanning receptors (7-TMR), which may reflect direct coupling of the GPR-Gαi module to the receptor analogous to canonical Gαßγ heterotrimer. We have demonstrated that the AGS4-Gαi complex is regulated by chemokine receptors in an agonist-dependent manner that is receptor-proximal. As an initial approach to investigate the functional role(s) of this regulated interaction in vivo, we analyzed leukocytes, in which AGS4/Gpsm3 is predominantly expressed, from AGS4/Gpsm3-null mice. Loss of AGS4/Gpsm3 resulted in mild but significant neutropenia and leukocytosis. Dendritic cells, T lymphocytes, and neutrophils from AGS4/Gpsm3-null mice also exhibited significant defects in chemoattractant-directed chemotaxis and extracellular signal-regulated kinase activation. An in vivo peritonitis model revealed a dramatic reduction in the ability of AGS4/Gpsm3-null neutrophils to migrate to primary sites of inflammation. Taken together, these data suggest that AGS4/Gpsm3 is required for proper chemokine signal processing in leukocytes and provide further evidence for the importance of the GPR-Gαi module in the regulation of leukocyte function.


Subject(s)
Chemokines/metabolism , Chemotaxis, Leukocyte/physiology , Dendritic Cells/physiology , Guanine Nucleotide Dissociation Inhibitors/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Neutrophils/physiology , T-Lymphocytes/physiology , Animals , Chemotactic Factors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , RGS Proteins/metabolism , Signal Transduction/physiology
6.
Mol Cell Oncol ; 3(5): e1078923, 2016.
Article in English | MEDLINE | ID: mdl-27857968

ABSTRACT

Paradoxically, both anticancer immunosurveillance and tumor progression have been associated with intact autophagy, which is regulated by the target of rapamycin (Tor1). Here, we describe the potential impact on the design of cancer therapeutics of a newly described highly conserved post-transcriptional mechanism whereby Tor regulates autophagy.

7.
J Immunol ; 196(2): 846-56, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26667172

ABSTRACT

Many intracellular pathogens cause disease by subverting macrophage innate immune defense mechanisms. Intracellular pathogens actively avoid delivery to or directly target lysosomes, the major intracellular degradative organelle. In this article, we demonstrate that activator of G-protein signaling 3 (AGS3), an LPS-inducible protein in macrophages, affects both lysosomal biogenesis and activity. AGS3 binds the Gi family of G proteins via its G-protein regulatory (GoLoco) motif, stabilizing the Gα subunit in its GDP-bound conformation. Elevated AGS3 levels in macrophages limited the activity of the mammalian target of rapamycin pathway, a sensor of cellular nutritional status. This triggered the nuclear translocation of transcription factor EB, a known activator of lysosomal gene transcription. In contrast, AGS3-deficient macrophages had increased mammalian target of rapamycin activity, reduced transcription factor EB activity, and a lower lysosomal mass. High levels of AGS3 in macrophages enhanced their resistance to infection by Burkholderia cenocepacia J2315, Mycobacterium tuberculosis, and methicillin-resistant Staphylococcus aureus, whereas AGS3-deficient macrophages were more susceptible. We conclude that LPS priming increases AGS3 levels, which enhances lysosomal function and increases the capacity of macrophages to eliminate intracellular pathogens.


Subject(s)
Bacterial Infections/immunology , Carrier Proteins/immunology , Lysosomes/immunology , Macrophages/immunology , Macrophages/microbiology , Animals , Flow Cytometry , Guanine Nucleotide Dissociation Inhibitors , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Polymerase Chain Reaction , RNA, Small Interfering
8.
Autophagy ; 11(12): 2390-2, 2015.
Article in English | MEDLINE | ID: mdl-26569496

ABSTRACT

Regulation of autophagy is required to maintain cellular equilibrium and prevent disease. While extensive study of post-translational mechanisms has yielded important insights into autophagy induction, less is known about post-transcriptional mechanisms that could potentiate homeostatic control. In our study, we showed that the RNA-binding protein, Dhh1 in Saccharomyces cerevisiae and Vad1 in the pathogenic yeast Cryptococcus neoformans is involved in recruitment and degradation of key autophagy mRNAs. In addition, phosphorylation of the decapping protein Dcp2 by the target of rapamycin (TOR), facilitates decapping and degradation of autophagy-related mRNAs, resulting in repression of autophagy under nutrient-replete conditions. The post-transcriptional regulatory process is conserved in both mouse and human cells and plays a role in autophagy-related modulation of the inflammasome product IL1B. These results were then applied to provide mechanistic insight into autoimmunity of a patient with a PIK3CD/p110δ gain-of-function mutation. These results thus identify an important new post-transcriptional mechanism of autophagy regulation that is highly conserved between yeast and mammals.


Subject(s)
Autophagy/genetics , DEAD-box RNA Helicases/genetics , RNA Stability/genetics , Saccharomyces cerevisiae Proteins/genetics , Animals , Female , Humans
9.
Nat Cell Biol ; 17(7): 930-942, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26098573

ABSTRACT

Autophagy is an essential eukaryotic pathway requiring tight regulation to maintain homeostasis and preclude disease. Using yeast and mammalian cells, we report a conserved mechanism of autophagy regulation by RNA helicase RCK family members in association with the decapping enzyme Dcp2. Under nutrient-replete conditions, Dcp2 undergoes TOR-dependent phosphorylation and associates with RCK members to form a complex with autophagy-related (ATG) mRNA transcripts, leading to decapping, degradation and autophagy suppression. Simultaneous with the induction of ATG mRNA synthesis, starvation reverses the process, facilitating ATG mRNA accumulation and autophagy induction. This conserved post-transcriptional mechanism modulates fungal virulence and the mammalian inflammasome, the latter providing mechanistic insight into autoimmunity reported in a patient with a PIK3CD/p110δ gain-of-function mutation. We propose a dynamic model wherein RCK family members, in conjunction with Dcp2, function in controlling ATG mRNA stability to govern autophagy, which in turn modulates vital cellular processes affecting inflammation and microbial pathogenesis.


Subject(s)
Autophagy/genetics , DEAD-box RNA Helicases/genetics , RNA Stability/genetics , Saccharomyces cerevisiae Proteins/genetics , Animals , Autoimmunity/genetics , Cell Line, Tumor , Cells, Cultured , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Female , Gene Expression Regulation, Fungal , HeLa Cells , Humans , Immunoblotting , Inflammasomes/genetics , Inflammasomes/metabolism , Mice, Inbred C57BL , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
10.
Kardiol Pol ; 73(9): 740-6, 2015.
Article in English | MEDLINE | ID: mdl-25985726

ABSTRACT

BACKGROUND: In recent years, many microRNAs (miRNAs) were shown to be dysregulated in specific tissues playing critical roles in the pathogenesis and progression of heart failure (HF). Left ventricular (LV) mass (LVM) has long been recognised as an important prognostic marker in systolic HF patients. AIM: We hypothesised that circulating miRNAs may be associated with LVM in systolic HF patients. The present study aimed to evaluate the relationship between previously reported and novel dysregulated circulating miRNAs and echocardiographically determined LVM in symptomatic HF patients with LV systolic dysfunction. METHODS: Forty-two consecutive patients diagnosed with NYHA II-IV symptomatic systolic HF and a control group consisting of 15 age- and sex-matched healthy volunteers were enrolled. After labelling extracted RNA, poly-A tails were added. RNAs were later hybridised on a GeneChip miRNA 2.0 array. After hybridisation and staining, arrays were scanned to determine miRNA expression levels, and differentially expressed miRNAs were identified. RESULTS: Eighteen miRNAs were found to be upregulated in serum of HF patients, while 11 were demonstrated to be downregulated. When the association between dysregulated miRNAs and echocardiographic findings was investigated, miR-182 (p = 0.04), miR-200a* (p = 0.019), and miR-568 (p = 0.023) were found to be inversely correlated with LVM index (LVMI), while miR-155 (p = 0.019) and miR-595 (p = 0.04) were determined to be positively correlated with LVMI. CONCLUSIONS: The results of our study revealed that dysregulated circulating miRNAs were correlated with anatomic changes in LV, in terms of LVMI, in symptomatic HF patients with systolic LV dysfunction.


Subject(s)
Heart Failure/complications , MicroRNAs/genetics , Ventricular Dysfunction, Left/complications , Adult , Aged , Echocardiography , Female , Gene Expression Regulation , Heart Failure/blood , Heart Failure/metabolism , Humans , Male , MicroRNAs/blood , Middle Aged , Systole , Ventricular Dysfunction, Left/metabolism
11.
J Cardiovasc Med (Hagerstown) ; 16(6): 431-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25643195

ABSTRACT

INTRODUCTION: Recent studies have demonstrated the potential of microRNAs (miRNA) as biomarkers in various cardiovascular disorders. The aim of the present study was to quantitatively evaluate the expression levels of miRNAs in patients with chronic congestive heart failure (CHF) in order to identify differential expression profiles as biomarkers with prognostic values. MATERIALS AND METHOD: The study included 20 clinically stable [New York Heart Association (NYHA) II] and 22 decompensated (NYHA III and IV) CHF patients and 15 healthy controls. miRNA profiling was performed using a microarray method. Dysregulated miRNAs were evaluated for their biomarker potential. RESULTS: Microarray profiling revealed an increase in the expression of miR-21, miR-650, miR-744, miR-516-5p, miR-1292, miR-182, miR-1228, miR-595, miR-663b, miR-1296, miR-1825, miR-299-3p, miR-662 miR-122, miR-3148 and miR-518e and a decrease in the expression of miR-129-3p, miR-3155, miR-3175, miR-583, miR-568, miR-30d, miR-200a-star, miR-1979, miR-371-3p, miR-155-star and miR-502-5p in sera of CHF patients. The prognostic value of miR-182 [area under the curve (AUC) 0.695] was found to be superior to pro-brain type natriuretic peptide (NT-proBNP; AUC 0.350) and high-sensitivity C-reactive protein (hs-CRP) (AUC 0.475) by receiver operator characteristic (ROC) analysis. Cox regression analysis showed that miR-182 could predict cardiovascular mortality (P = 0.032). CONCLUSION: We demonstrated the increased expression levels of circulating miRNAs in CHF as compared with controls. Moreover, miR-182 was found to be a potential prognostic marker in CHF.


Subject(s)
Heart Failure/diagnosis , MicroRNAs/blood , Adult , Aged , Biomarkers/blood , C-Reactive Protein/analysis , Case-Control Studies , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Genome-Wide Association Study/methods , Heart Failure/genetics , Humans , Male , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Prognosis , Prospective Studies
12.
Anatol J Cardiol ; 15(9): 716-21, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25592107

ABSTRACT

OBJECTIVE: Apelin is a novel endogenous peptide with inotropic and vasodilatory properties and is the ligand for the angiotensin receptor-like 1 (APJ) receptor. The aim of the study was to investigate the association of 2 single-nucleotide polymorphisms (SNPs) in the apelin gene with susceptibility to coronary artery disease (CAD) in the Turkish population. METHODS: The present observational case-control study consisted of 244 subjects (134 angiographically proven CAD patients and 110 healthy controls) aged 30-65 years. The association of 2 SNPs (rs3115758 and rs3115759) in the apelin gene and CAD risk was investigated. Real-time polymerase chain reaction (RT-PCR) was used to analyze the 2 SNPs in both the CAD and the healthy subjects. Allele and genotype frequencies between patients and control groups were compared using the Chi-square (χ2) test. The relationships of the 2 polymorphisms with the presence of CAD were determined with multiple binary logistic regression analysis after adjustment for CAD risk factors. RESULTS: TT and AA risk genotypes of the rs3115758 and rs3115759 variants in the apelin gene were found to be significantly related with the risk of CAD with the same power (OR: 6.36, 95% CI: 1.41-28.6) (p=0.007). After adjustments for traditional CAD risk factors, the homozygous TT genotype for rs3115758 and AA genotype for rs3115759 increased the CAD risk, both with an OR of 5.91. CONCLUSION: Genetic variants in the apelin gene are significantly associated with the risk of CAD in the Turkish population.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Coronary Artery Disease/genetics , Receptors, G-Protein-Coupled/genetics , Adult , Aged , Apelin Receptors , Case-Control Studies , Coronary Artery Disease/mortality , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Turkey , White People/genetics
13.
Microb Cell ; 2(8): 302-304, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-28357306

ABSTRACT

Eukaryotic cells utilize macroautophagy (hereafter autophagy) to recycle cellular materials during nutrient stress. Target of rapamycin (Tor) is a central regulator of this process, acting by post-translational mechanisms, phosphorylating preformed autophagy-related (Atg) proteins to repress autophagy during log-phase growth. We recently reported an additional role for post-transcriptional regulation of autophagy, whereby the mRNA decapping protein, Dcp2, undergoes Tor-dependent phosphorylation, resulting in increased ATG mRNA decapping and degradation under nutrient-rich, repressing conditions. Dephosphorylation of Dcp2 during starvation is associated with dissociation of the decapping-ATG mRNA complex, with resultant stabilization of, and accumulation of, ATG transcripts, leading to induction of autophagy. Regulation of mRNA degradation occurs in concert with known mRNA synthetic inductive mechanisms to potentiate overall transcriptional regulation. This mRNA degradative pathway thus constitutes a type of transcriptional 'futile cycle' where under nutrient-rich conditions transcript is constantly being generated and degraded. As nutrient levels decline, steady state mRNA levels are increased by both inhibition of degradation as well as increased de novo synthesis. A role for this regulatory process in fungal virulence was further demonstrated by showing that overexpression of the Dcp2-associated mRNA-binding protein Vad1 in the AIDS-associated pathogen Cryptococcus neoformans results in constitutive repression of autophagy even under starvation conditions as well as attenuated virulence in a mouse model. In summary, Tor-dependent post-transcriptional regulation of autophagy plays a key role in the facilitation of microbial pathogenesis.

14.
Anatol J Cardiol ; 15(3): 196-203, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25333979

ABSTRACT

OBJECTIVE: Coronary artery disease (CAD), which develops from complex interactions between genetic and enviromental factors, is a leading cause of death worldwide. Based on genome-wide association studies (GWAS), the chromosomal region 9p21 has been identified as the most relevant locus presenting a strong association with CAD in different populations. The aim of the present study was to investigate the association of two SNPs on chromosome 9p21 on susceptibility to CAD and the effect of these SNPs along with cardiovascular risk factors on the severity of CAD in the Turkish population. METHODS: This study had an observational case-control design. We genotyped 460 subjects, aged 30-65 years, to investigate the association of 2 SNPs (rs1333049, rs2383207) on chromosome 9p21 and CAD risk in Turkish population. Real-time polymerase chain reaction (RT-PCR) was used to analyze the 2 SNPs in CAD patients and healthy controls. The genotype and allelic variations of these SNPs with the severity of CAD was also assessed using semi-quantitative methods such as the Gensini score. Student's t test and multiple regression analysis were used for statistical analysis. RESULTS: The SNPs rs1333049 and rs2383207 were found to be associated with CAD with an adjusted OR of 1.81 (95% Cl 1.05-3.12) and 2.12 (95% CI 1.19-4.10) respectively. After adjustment of CAD risk factors such as smoking, family history of CAD and diabetes, the homozygous AA genotype for rs2383207 increased the CAD risk with an OR 3.69. Also a very strong association was found between rs1333049 and rs2383207 and Gensini scores representing the severity of CAD (p<0.001). CONCLUSION: The rs2383207 and rs1333049 SNPs on 9p21 chromosome were significantly associated with the risk and severity of CAD in the Turkish population.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Adult , Aged , Case-Control Studies , Coronary Artery Disease/pathology , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction , Severity of Illness Index , Turkey , White People/genetics
15.
Mol Cell Biol ; 34(22): 4186-99, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25225330

ABSTRACT

Both chemotaxis and phagocytosis depend upon actin-driven cell protrusions and cell membrane remodeling. While chemoattractant receptors rely upon canonical G-protein signaling to activate downstream effectors, whether such signaling pathways affect phagocytosis is contentious. Here, we report that Gαi nucleotide exchange and signaling helps macrophages coordinate the recognition, capture, and engulfment of zymosan bioparticles. We show that zymosan exposure recruits F-actin, Gαi proteins, and Elmo1 to phagocytic cups and early phagosomes. Zymosan triggered an increase in intracellular Ca(2+) that was partially sensitive to Gαi nucleotide exchange inhibition and expression of GTP-bound Gαi recruited Elmo1 to the plasma membrane. Reducing GDP-Gαi nucleotide exchange, decreasing Gαi expression, pharmacologically interrupting Gßγ signaling, or reducing Elmo1 expression all impaired phagocytosis, while favoring the duration that Gαi remained GTP bound promoted it. Our studies demonstrate that targeting heterotrimeric G-protein signaling offers opportunities to enhance or retard macrophage engulfment of phagocytic targets such as zymosan.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/immunology , Macrophages/cytology , Phagocytosis , Signal Transduction , Zymosan/immunology , Actins/analysis , Actins/immunology , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/immunology , Animals , Calcium/analysis , Calcium/immunology , Cell Line , GTP-Binding Protein alpha Subunit, Gi2/analysis , GTP-Binding Protein alpha Subunit, Gi2/genetics , Gene Deletion , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Phagosomes/genetics , Phagosomes/immunology , Phagosomes/microbiology , Phagosomes/ultrastructure , Saccharomyces cerevisiae/immunology
16.
PLoS One ; 9(6): e97957, 2014.
Article in English | MEDLINE | ID: mdl-24911523

ABSTRACT

The omega-3 (ω3) fatty acid docosahexaenoic acid (DHA) can suppress inflammation, specifically IL-1ß production through poorly understood molecular mechanisms. Here, we show that DHA reduces macrophage IL-1ß production by limiting inflammasome activation. Exposure to DHA reduced IL-1ß production by ligands that stimulate the NLRP3, AIM2, and NAIP5/NLRC4 inflammasomes. The inhibition required Free Fatty Acid Receptor (FFAR) 4 (also known as GPR120), a G-protein coupled receptor (GPR) known to bind DHA. The exposure of cells to DHA recruited the adapter protein ß-arrestin1/2 to FFAR4, but not to a related lipid receptor. DHA treatment reduced the initial inflammasome priming step by suppressing the nuclear translocation of NF-κB. DHA also reduced IL-1ß levels by enhancing autophagy in the cells. As a consequence macrophages derived from mice lacking the essential autophagy protein ATG7 were partially resistant to suppressive effects of DHA. Thus, DHA suppresses inflammasome activation by two distinct mechanisms, inhibiting the initial priming step and by augmenting autophagy, which limits inflammasome activity.


Subject(s)
Autophagy/drug effects , Docosahexaenoic Acids/pharmacology , Inflammasomes/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , NF-kappa B/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Arrestins/metabolism , Bone Marrow Cells/cytology , Calcium/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Intracellular Space/drug effects , Intracellular Space/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/genetics , beta-Arrestins
17.
Scientifica (Cairo) ; 2014: 825463, 2014.
Article in English | MEDLINE | ID: mdl-24818040

ABSTRACT

Macrophages are on the front line of host defense. They possess an array of germline-encoded pattern recognition receptors/sensors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) and which activate downstream effectors/pathways to help mediate innate immune responses and host defense. Innate immune responses include the rapid induction of transcriptional networks that trigger the production of cytokines, chemokines, and cytotoxic molecules; the mobilization of cells including neutrophils and other leukocytes; the engulfment of pathogens by phagocytosis and their delivery to lysosome for degradation; and the induction of autophagy. Autophagy is a catabolic process that normally maintains cellular homeostasis in a lysosome-dependent manner, but it also functions as a cytoprotective response that intersects with a variety of general stress-response pathways. This review focuses on the intimately linked molecular mechanisms that help govern the autophagic pathway and macrophage innate immune responses.

18.
PLoS One ; 8(11): e81886, 2013.
Article in English | MEDLINE | ID: mdl-24312373

ABSTRACT

In macrophages autophagy assists antigen presentation, affects cytokine release, and promotes intracellular pathogen elimination. In some cells autophagy is modulated by a signaling pathway that employs Gαi3, Activator of G-protein Signaling-3 (AGS3/GPSM1), and Regulator of G-protein Signaling 19 (RGS19). As macrophages express each of these proteins, we tested their importance in regulating macrophage autophagy. We assessed LC3 processing and the formation of LC3 puncta in bone marrow derived macrophages prepared from wild type, Gnai3(-/-), Gpsm1(-/-), or Rgs19(-/-) mice following amino acid starvation or Nigericin treatment. In addition, we evaluated rapamycin-induced autophagic proteolysis rates by long-lived protein degradation assays and anti-autophagic action after rapamycin induction in wild type, Gnai3(-/-), and Gpsm1(-/-) macrophages. In similar assays we compared macrophages treated or not with pertussis toxin, an inhibitor of GPCR (G-protein couple receptor) triggered Gαi nucleotide exchange. Despite previous findings, the level of basal autophagy, autophagic induction, autophagic flux, autophagic degradation and the anti-autophagic action in macrophages that lacked Gαi3, AGS3, or RGS19; or had been treated with pertussis toxin, were similar to controls. These results indicate that while Gαi signaling may impact autophagy in some cell types it does not in macrophages.


Subject(s)
Autophagy , Carrier Proteins/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Macrophages/immunology , RGS Proteins/physiology , Animals , Carrier Proteins/genetics , Cells, Cultured , Flow Cytometry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Guanine Nucleotide Dissociation Inhibitors , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Proteolysis , RGS Proteins/genetics
19.
J Biol Chem ; 288(33): 24091-103, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23770668

ABSTRACT

Group II activators of G-protein signaling play diverse functional roles through their interaction with Gαi, Gαt, and Gαo via a G-protein regulatory (GPR) motif that serves as a docking site for Gα-GDP. We recently reported the regulation of the AGS3-Gαi signaling module by a cell surface, seven-transmembrane receptor. Upon receptor activation, AGS3 reversibly dissociates from the cell cortex, suggesting that it may function as a signal transducer with downstream signaling implications, and this question is addressed in the current report. In HEK-293 and COS-7 cells expressing the α2A/D-AR and Gαi3, receptor activation resulted in the translocation of endogenous AGS3 and AGS3-GFP from the cell cortex to a juxtanuclear region, where it co-localized with markers of the Golgi apparatus (GA). The agonist-induced translocation of AGS3 was reversed by the α2-AR antagonist rauwolscine. The TPR domain of AGS3 was required for agonist-induced translocation of AGS3 from the cell cortex to the GA, and the translocation was blocked by pertussis toxin pretreatment or by the phospholipase Cß inhibitor U73122. Agonist-induced translocation of AGS3 to the GA altered the functional organization and protein sorting at the trans-Golgi network. The regulated movement of AGS3 between the cell cortex and the GA offers unexpected mechanisms for modulating protein secretion and/or endosome recycling events at the trans-Golgi network.


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/metabolism , Receptors, Cell Surface/metabolism , trans-Golgi Network/metabolism , Animals , Biomarkers/metabolism , Brimonidine Tartrate , COS Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , Endosomes/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Green Fluorescent Proteins/metabolism , Guanine Nucleotide Dissociation Inhibitors/chemistry , HEK293 Cells , Humans , Lysosomes/metabolism , Protein Structure, Tertiary , Protein Transport/drug effects , Quinoxalines/pharmacology , Subcellular Fractions/metabolism , Time Factors , trans-Golgi Network/drug effects
20.
Mol Cell Biochem ; 379(1-2): 77-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23535969

ABSTRACT

Metabolic syndrome (MetS) is a common multifactorial disorder that involves abdominal obesity, dyslipidemia, hypertension, and hyperglycemia. Genome-wide association studies have identified a major risk locus for coronary artery disease and myocardial infarction on chromosome 9p21. Here, we examined the frequency of single nucleotide polymorphisms (SNPs) on chromosome 9p21 in a sample of Turkish patients with MetS and further investigated the correlation between regional SNPs, haplotypes, and MetS. The real-time polymerase chain reaction (RT-PCR) was used to analyze 4 SNPs (rs10757274 A/G, rs2383207 A/G, rs10757278 A/G, rs1333049 C/G) in 291 MetS patients and 247 controls. Analysis of 4 SNPs revealed a significant difference in the genotype distribution for rs2383207, rs10757278, and rs1333049 between MetS patients and controls (p = 0.041, p = 0.005, p = 0.023, respectively) but not for rs10757274 (p = 0.211). MetS and control allelic frequencies for rs2383207, rs10757278, and rs1333049 were statistically different (p < 0.05). The rs2383207 AG variant, was identified as a MetS risk factor (p = 0.012, OR = 33.271; 95 % CI: 2.193-504.805) and the AA haplotype in block 1 and the GC, AG haplotypes in block 2 were associated with MetS (χ(2) = 3.875, p = 0.049; χ(2) = 9.334, p = 0.0022; χ (2) = 9.134, p = 0.0025, respectively). In this study, we found that chromosome 9p21 SNP rs10757278 and related haplotypes correlate with MetS risk. This is the first report showing an association between a 9p21 variant and MetS and suggests that rs10757278 polymorphism may confer increased risk for disease.


Subject(s)
Chromosomes, Human, Pair 9/genetics , Metabolic Syndrome/genetics , Polymorphism, Single Nucleotide , Case-Control Studies , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Linkage Disequilibrium , Male , Middle Aged , Risk Factors , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...