Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
J Immunol ; 192(5): 2186-94, 2014 Mar 01.
Article En | MEDLINE | ID: mdl-24510965

The IL-8 (CXCL8) receptors CXCR1 and CXCR2 couple to Gαi to induce leukocyte recruitment and activation at sites of inflammation. We recently showed that CXCR1 couples predominantly to the G protein-coupled receptor kinase (GRK)2, whereas CXCR2 interacts with GRK6 to regulate cellular responses. In addition to G protein-coupled receptors, GRKs displayed a more diverse protein/protein interaction in cells. In this study, we sought to identify GRK6 binding partner(s) that may influence CXCL8 activities, using RBL-2H3 cells stably expressing CXCR1 (RBL-CXCR1) or CXCR2 (RBL-CXCR2), as well as human and murine neutrophils. Our data demonstrated that, upon CXCR2 activation, GRK6 interacts with activator of G protein signaling (AGS)3 and Gαi2 to form a GRK6/AGS3/Gαi2 complex. This complex is time dependent and peaked at 2-3 min postactivation. GTPγS pretreatment blocked GRK6/AGS3/Gαi2 formation, suggesting that this assembly depends on G protein activation. Surprisingly, CXCR2 activation induced AGS3 phosphorylation in a PKC-dependent, but GRK6-independent, fashion. Overexpression of AGS3 in RBL-CXCR2 significantly inhibited CXCL8-induced Ca(2+) mobilization, phosphoinositide hydrolysis, and chemotaxis. In contrast, short hairpin RNA inhibition of AGS3 enhanced CXCL8-induced Ca(2+) mobilization, receptor resistance to desensitization, and recycling to the cell surface, with no effect on receptor internalization. Interestingly, RBL-CXCR2-AGS3(-/-) cells displayed a significant increase in CXCR2 expression on the cell surface but decreased ERK1/2 and P38 MAPK activation. Taken together, these results indicate that GRK6 complexes with AGS3-Gαi2 to regulate CXCR2-mediated leukocyte functions at different levels, including downstream effector activation, receptor trafficking, and expression at the cell membrane.


G-Protein-Coupled Receptor Kinases/immunology , Guanine Nucleotide Dissociation Inhibitors/immunology , Multiprotein Complexes/immunology , Receptors, Interleukin-8B/immunology , Animals , Calcium/immunology , Calcium Signaling/genetics , Calcium Signaling/immunology , Cell Line , Cell Membrane/genetics , Cell Membrane/immunology , G-Protein-Coupled Receptor Kinases/genetics , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/immunology , Gene Expression Regulation/physiology , Guanine Nucleotide Dissociation Inhibitors/genetics , Humans , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Phosphorylation/genetics , Phosphorylation/immunology , Protein Transport/physiology , Receptors, Interleukin-8B/genetics
2.
J Immunol ; 190(10): 5329-36, 2013 May 15.
Article En | MEDLINE | ID: mdl-23589623

G protein-coupled receptor kinases (GRKs) phosphorylate the activated form of G protein-coupled receptors leading to receptor desensitization and downregulation. We have recently shown that the chemokine receptor, CXCR2, couples to GRK6 to regulate cellular responses including chemotaxis, angiogenesis, and wound healing. In this study, we investigate the role of GRK6 in tumorigenesis using murine models of human lung cancer. Mice deficient in GRK6 (GRK6(-/-)) exhibited a significant increase in Lewis lung cancer growth and metastasis relative to control littermates (GRK6(+/+)). GRK6 deletion had no effect on the expression of proangiogenic chemokine or vascular endothelial growth factor, but upregulated matrix metalloproteinase (MMP)-2 and MMP-9 release, tumor-infiltrating PMNs, and microvessel density. Because ß-arrestin-2-deficient (ßarr2(-/-)) mice exhibited increased Lewis lung cancer growth and metastasis similar to that of GRK6(-/-), we developed a double GRK6(-/-)/ßarr2(-/-) mouse model. Surprisingly, GRK6(-/-)/ßarr2(-/-) mice exhibited faster tumor growth relative to GRK6(-/-) or ßarr2(-/-) mice. Treatment of the mice with anti-CXCR2 Ab inhibited tumor growth in both GRK6(-/-) and GRK6(-/-)/ßarr2(-/-) animals. Altogether, the results indicate that CXCR2 couples to GRK6 to regulate angiogenesis, tumor progression, and metastasis. Deletion of GRK6 increases the activity of the host CXCR2, resulting in greater PMN infiltration and MMP release in the tumor microenvironment, thereby promoting angiogenesis and metastasis. Because GRK6(-/-)/ßarr2(-/-) showed greater tumor growth relative to GRK6(-/-) or ßarr2(-/-) mice, the data further suggest that CXCR2 couples to different mechanisms to mediate tumor progression and metastasis.


Arrestins/genetics , Carcinoma, Lewis Lung/metabolism , G-Protein-Coupled Receptor Kinases/genetics , G-Protein-Coupled Receptor Kinases/metabolism , Neutrophils/immunology , Receptors, Interleukin-8B/metabolism , Animals , Arrestins/deficiency , Arrestins/metabolism , Cell Line, Tumor , Chemotaxis , Disease Progression , Down-Regulation , G-Protein-Coupled Receptor Kinases/deficiency , Genotype , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Knockout , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phenylurea Compounds/pharmacology , Phosphorylation , Receptors, Interleukin-8B/antagonists & inhibitors , Signal Transduction , Tumor Microenvironment , Up-Regulation , Wound Healing , beta-Arrestin 2 , beta-Arrestins
3.
J Immunol ; 189(6): 2824-32, 2012 Sep 15.
Article En | MEDLINE | ID: mdl-22869904

The chemokine receptors, CXCR1 and CXCR2, couple to Gαi to induce leukocyte recruitment and activation at sites of inflammation. Upon activation by CXCL8, these receptors become phosphorylated, desensitized, and internalized. In this study, we investigated the role of different G protein-coupled receptor kinases (GRKs) in CXCR1- and CXCR2-mediated cellular functions. To that end, short hairpin RNA was used to inhibit GRK2, 3, 5, and 6 in RBL-2H3 cells stably expressing CXCR1 or CXCR2, and CXCL8-mediated receptor activation and regulation were assessed. Inhibition of GRK2 and GRK6 increased CXCR1 and CXCR2 resistance to phosphorylation, desensitization, and internalization, respectively, and enhanced CXCL8-induced phosphoinositide hydrolysis and exocytosis in vitro. GRK2 depletion diminished CXCR1-induced ERK1/2 phosphorylation but had no effect on CXCR2-induced ERK1/2 phosphorylation. GRK6 depletion had no significant effect on CXCR1 function. However, peritoneal neutrophils from mice deficient in GRK6 (GRK6(-/-)) displayed an increase in CXCR2-mediated G protein activation but in vitro exhibited a decrease in chemotaxis, receptor desensitization, and internalization relative to wild-type (GRK6(+/+)) cells. In contrast, neutrophil recruitment in vivo in GRK6(-/-) mice was increased in response to delivery of CXCL1 through the air pouch model. In a wound-closure assay, GRK6(-/-) mice showed enhanced myeloperoxidase activity, suggesting enhanced neutrophil recruitment, and faster wound closure compared with GRK6(+/+) animals. Taken together, the results indicate that CXCR1 and CXCR2 couple to distinct GRK isoforms to mediate and regulate inflammatory responses. CXCR1 predominantly couples to GRK2, whereas CXCR2 interacts with GRK6 to negatively regulate receptor sensitization and trafficking, thus affecting cell signaling and angiogenesis.


G-Protein-Coupled Receptor Kinases/metabolism , Neutrophils/immunology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Cell Line, Tumor , Exocytosis/genetics , Exocytosis/immunology , Female , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 2/deficiency , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinases/antagonists & inhibitors , G-Protein-Coupled Receptor Kinases/deficiency , Humans , Interleukin-8/physiology , Leukemia, Basophilic, Acute/genetics , Leukemia, Basophilic, Acute/immunology , Leukemia, Basophilic, Acute/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/immunology , Neutrophils/enzymology , Neutrophils/metabolism , Phosphorylation/genetics , Rats , Receptors, Interleukin-8A/physiology , Receptors, Interleukin-8B/physiology , Signal Transduction/genetics , Signal Transduction/immunology
4.
Blood ; 118(20): 5528-39, 2011 Nov 17.
Article En | MEDLINE | ID: mdl-21849482

Endoplasmic reticulum (ER) stress triggers a homeostatic cellular response in mammalian cells to ensure efficient folding, sorting, and processing of client proteins. In lytic-permissive lymphoblastoid cell lines (LCLs), pulse exposure to the chemical ER-stress inducer thapsigargin (TG) followed by recovery resulted in the activation of the EBV immediate-early (BRLF1, BZLF1), early (BMRF1), and late (gp350) genes, gp350 surface expression, and virus release. The protein phosphatase 1 a (PP1a)-specific phosphatase inhibitor Salubrinal (SAL) synergized with TG to induce EBV lytic genes; however, TG treatment alone was sufficient to activate EBV lytic replication. SAL showed ER-stress-dependent and -independent antiviral effects, preventing virus release in human LCLs and abrogating gp350 expression in 12-O-tetradecanoylphorbol-13-acetate (TPA)-treated B95-8 cells. TG resulted in sustained BCL6 but not BLIMP1 or CD138 expression, which is consistent with maintenance of a germinal center B-cell, rather than plasma-cell, phenotype. Microarray analysis identified candidate genes governing lytic replication in LCLs undergoing ER stress.


Endoplasmic Reticulum Stress/physiology , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/genetics , Lymphoma/virology , Virus Replication/physiology , Carcinogens/pharmacology , Cell Line , Cinnamates/pharmacology , Enzyme Inhibitors/pharmacology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/physiopathology , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling , Gene Expression Regulation, Viral/physiology , Genes, Immediate-Early/genetics , Germinal Center/cytology , Germinal Center/metabolism , Germinal Center/virology , Herpesvirus 4, Human/drug effects , Humans , Immediate-Early Proteins/genetics , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphocytes/virology , Lymphoma/metabolism , Membrane Glycoproteins/genetics , Plasma Cells/cytology , Plasma Cells/metabolism , Plasma Cells/virology , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Trans-Activators/genetics , Viral Matrix Proteins/genetics , Virus Replication/drug effects
5.
J Immunol ; 183(5): 3425-32, 2009 Sep 01.
Article En | MEDLINE | ID: mdl-19667085

CXCL8 (also known as IL-8) activates CXCR1 and CXCR2 to mediate neutrophil recruitment and trigger cytotoxic effect at sites of infection. Under physiological conditions, CXCL8 could exist as monomers, dimers, or a mixture of monomers and dimers. Therefore, both forms of CXCL8 could interact with CXCR1 and CXCR2 with different affinities and potencies to mediate different cellular responses. In the present study, we have used a "trapped" nonassociating monomer (L25NMe) and a nondissociating dimer (R26C) to investigate their activities for human neutrophils that express both receptors and for RBL-2H3 cells stably expressing either CXCR1(RBL-CXCR1) or CXCR2 (RBL-CXCR2). The monomer was more active than the dimer for activities such as intracellular Ca(2+) mobilization, phosphoinositide hydrolysis, chemotaxis. and exocytosis. Receptor regulation, however, is distinct for each receptor. The rate of monomer-mediated regulation of CXCR1 is greater for activities such as phosphorylation, desensitization, beta-arrestin translocation, and internalization. In contrast, for CXCR2, both monomeric and dimeric CXCL8 mediate these activities to a similar extent. Interestingly, receptor-mediated signal-regulated kinase (ERK) phosphorylation in response to all three CXCL8 variants was more sustained for CXCR2 relative to CXCR1. Taken together, the results indicate that the CXCL8 monomer and dimer differentially activate and regulate CXCR1 and CXCR2 receptors. These distinct properties of the ligand and the receptors play a critical role in orchestrating neutrophil recruitment and eliciting cytotoxic activity during an inflammatory response.


Interleukin-8/chemistry , Interleukin-8/physiology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Cell Line , Cell Line, Tumor , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Dimerization , Gene Expression Regulation/immunology , Humans , Inflammation Mediators/chemistry , Inflammation Mediators/physiology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Rats , Receptors, Interleukin-8A/biosynthesis , Receptors, Interleukin-8A/chemistry , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8B/biosynthesis , Receptors, Interleukin-8B/chemistry , Receptors, Interleukin-8B/genetics
6.
Int Immunol ; 21(8): 991-1001, 2009 Aug.
Article En | MEDLINE | ID: mdl-19592420

Chemokine receptors (CCRs) are important co-stimulatory molecules found on many blood cells and associated with various diseases. The expression and function of CCRs on mast cells has been quite controversial. In this study, we report for the first time that murine bone marrow-derived mast cells (BMMC) express messenger RNA and protein for CCR1. BMMC cultured in the presence of murine recombinant stem cell factor and murine IL-3 expressed CCR1 after 5-6 weeks. We also report for the first time that mBMMC(CCR1+) cells endogenously express neurokinin receptor-1 and intercellular adhesion molecule-1. To examine the activity of CCR1 on these BMMC, we simultaneously stimulated two receptors: CCR1 by its ligand macrophage inflammatory protein-1alpha and the IgE receptor FcepsilonRI by antigen cross-linking. We found that co-stimulation enhanced BMMC degranulation compared with FcepsilonRI stimulation alone, as assessed by beta-hexosaminidase activity (85 versus 54%, P < 0.0001) and Ca(2+) influx (223 versus 183 nM, P < 0.05). We also observed significant increases in mast cell secretion of key growth factors, cytokines and chemokine mediators upon CCR1-FcepsilonRI co-stimulation. These factors include transforming growth factor beta-1, tumor necrosis factor-alpha and the cytokine IL-6. Taken together, our data indicate that CCR1 plays a key role in BMMC function. These findings contribute to our understanding of mechanisms for immune cell trafficking during inflammation.


Bone Marrow Cells/metabolism , Interleukin-6/metabolism , Mast Cells/metabolism , Receptors, CCR1/biosynthesis , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Female , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred BALB C , Receptors, CCR1/genetics , Receptors, CCR1/metabolism , Receptors, IgE/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction
7.
J Immunol ; 180(8): 5699-706, 2008 Apr 15.
Article En | MEDLINE | ID: mdl-18390755

Arrestins are adaptor/scaffold proteins that complex with activated and phosphorylated G protein-coupled receptor to terminate G protein activation and signal transduction. These complexes also mediate downstream signaling, independently of G protein activation. We have previously shown that beta-arrestin-2 (betaarr2) depletion promotes CXCR2-mediated cellular signaling, including angiogenesis and excisional wound closure. This study was designed to investigate the role of betaarr2 in tumorigenesis using a murine model of lung cancer. To that end, heterotopic murine Lewis lung cancer and tail vein metastasis tumor model systems in betaarr2-deficient mice (betaarr2(-/-)) and control littermates (betaarr2(+/+)) were used. betaarr2(-/-) mice exhibited a significant increase in Lewis lung cancer tumor growth and metastasis relative to betaarr2(+/+) mice. This correlated with decreased number of tumor-infiltrating lymphocytes but with elevated levels of the ELR(+) chemokines (CXCL1/keratinocyte-derived chemokine and CXCL2/MIP-2), vascular endothelial growth factor, and microvessel density. NF-kappaB activity was also enhanced in betaarr2(-/-) mice, whereas hypoxia-inducible factor-1alpha expression was decreased. Inhibition of CXCR2 or NF-kappaB reduced tumor growth in both betaarr2(-/-) and betaarr2(+/+) mice. NF-kappaB inhibition also decreased ELR(+) chemokines and vascular endothelial growth factor expression. Altogether, the data suggest that betaarr2 modulates tumorigenesis by regulating inflammation and angiogenesis through activation of CXCR2 and NF-kappaB.


Arrestins/metabolism , Chemokines, CXC/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , NF-kappa B/metabolism , Neovascularization, Pathologic , Receptors, Interleukin-8B/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Arrestins/deficiency , Arrestins/immunology , Chemokines, CXC/immunology , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/immunology , Mice , Mice, Mutant Strains , Receptors, Interleukin-8B/immunology , beta-Arrestin 2 , beta-Arrestins
8.
J Biol Chem ; 282(9): 6906-15, 2007 Mar 02.
Article En | MEDLINE | ID: mdl-17204468

CXCL8 (interleukin-8) interacts with two receptors, CXCR1 and CXCR2, to activate leukocytes. Upon activation, CXCR2 internalizes very rapidly relative to CXCR1 ( approximately 90% versus approximately 10% after 5 min). The C termini of the receptors have been shown to be necessary for internalization but are not sufficient to explain the distinct kinetics of down-regulation. To determine the structural determinant(s) that modulate receptor internalization, various chimeric and point mutant receptors were generated by progressively exchanging specific domains or amino acids between CXCR1 and CXCR2. The receptors were stably expressed in rat basophilic leukemia 2H3 cells and characterized for receptor binding, intracellular Ca(2+) mobilization, phosphoinositide hydrolysis, phosphorylation, internalization, and MAPK activation. The data herein indicate that the second extracellular loop (2ECL) of the receptors is critical for the distinct rate of internalization. Replacing the 2ECL of CXCR2 with that of CXCR1 (B(2ECL)A) or Asp(199) with its CXCR1 valine counterpart (B(D199V)A) delayed CXCR2 internalization similarly to CXCR1. Replacing Asp(199) with Asn (B(D199N)) restored CXCR2 rapid internalization. Structure modeling of the 2ECL of the receptors also suggested that Asp(199) plays a critical role in stabilizing and modulating CXCR2 rapid internalization relative to CXCR1. B(D199N) internalized rapidly but migrated as a single phosphorylated form like CXCR1 ( approximately 75 kDa), whereas B(2ECL)A and B(D199V)A showed slow and fast migrating forms like CXCR2 ( approximately 45 and approximately 65 kDa, respectively) but internalized like CXCR1. These data further undermine the role of receptor oligomerization in CXCL8 receptor internalization. Like CXCR1, B(D199V)A also induced sustained ERK activation and cross-desensitized Ca(2+) mobilization to CCR5 relative to B(D199N) and CXCR2. Altogether, the data suggest that the 2ECL of the CXCL8 receptors is important in modulating their distinct rate of down-regulation and thereby signal length and post-internalization activities.


Endocytosis , Interleukin-8/metabolism , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Aspartic Acid , Calcium Signaling , Cell Line, Tumor , Humans , Kinetics , Mutagenesis , Protein Structure, Secondary , Rats , Transfection
9.
J Immunol ; 177(5): 3242-9, 2006 Sep 01.
Article En | MEDLINE | ID: mdl-16920964

Platelet-activating factor (1-O-alkyl-2-acetyl-sn-glycerolphosphocholine; PAF) induces leukocyte accumulation and activation at sites of inflammation via the activation of a specific cell surface receptor (PAFR). PAFR couples to both pertussis toxin-sensitive and pertussis toxin-insensitive G proteins to activate leukocytes. To define the role(s) of G(i) and G(q) in PAF-induced leukocyte responses, two G-protein-linked receptors were generated by fusing G alpha(i3) (PAFR-G alpha(i3)) or G alpha(q) (PAFR-G alpha(q)) at the C terminus of PAFR. Rat basophilic leukemia cell line (RBL-2H3) stably expressing wild-type PAFR, PAFR-G alpha(i3), or PAFR-G alpha(q) was generated and characterized. All receptor variants bound PAF with similar affinities to mediate G-protein activation, intracellular Ca2+ mobilization, phosphoinositide (PI) hydrolysis, and secretion of beta-hexosaminidase. PAFR-G alpha(i3) and PAFR-G alpha(q) mediated greater GTPase activity in isolated membranes than PAFR but lower PI hydrolysis and secretion in whole cells. PAFR and PAFR-G alpha(i3), but not PAFR-G alpha(q), mediated chemotaxis to PAF. All three receptors underwent phosphorylation and desensitization upon exposure to PAF but only PAFR translocated beta arrestin to the cell membrane and internalized. In RBL-2H3 cells coexpressing the PAFRs along with CXCR1, IL-8 (CXCL8) cross-desensitized Ca2+ mobilization to PAF by all the receptors but only PAFR-G alpha(i3) activation cross-inhibited the response of CXCR1 to CXCL8. Altogether, the data indicate that G(i) exclusively mediates chemotactic and cross-regulatory signals of the PAFR, but both G(i) and G(q) activate PI hydrolysis and exocytosis by this receptor. Because chemotaxis and cross-desensitization are exclusively mediated by G(i), the data suggest that differential activation of both G(i) and G(q) by PAFR likely mediate specific as well as redundant signaling pathways.


Chemotaxis , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Calcium/metabolism , Cell Line, Tumor , Enzyme Activation , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Platelet Activating Factor/pharmacology , Protein Binding , Rats , Receptors, Interleukin-8A/metabolism , Signal Transduction/drug effects
10.
J Immunol ; 175(8): 5396-402, 2005 Oct 15.
Article En | MEDLINE | ID: mdl-16210646

CXCR2 is a G-protein-coupled receptor (GPCR) that binds the CXC chemokines, CXCL1-3 and CXCL5-8, and induces intracellular signals associated with chemotaxis. Many adaptor proteins are actively involved in the sequestration, internalization, and trafficking of CXCR2 and transduction of agonist-induced intracellular signaling. We have previously shown that adaptor protein beta-arrestin-2 (betaarr2) plays a crucial role in transducing signals mediated through CXCR2. To further investigate the role of betaarr2 on CXCR2-mediated signaling during acute inflammation, zymosan-induced neutrophils were isolated from peritoneal cavities of betaarr2-deficient (betaarr2(-/-)) and their wild-type (betaarr2(+/+)) littermate mice, and neutrophil CXCR2 signaling activities were determined by measurement of Ca(2+) mobilization, receptor internalization, GTPase activity, and superoxide anion production. The results showed that the deletion of betaarr2 resulted in increased Ca(2+) mobilization, superoxide anion production, and GTPase activity in neutrophils, but decreased receptor internalization relative to wild-type mice. Two animal models, the dorsal air pouch model and the excisional wound healing model, were used to further study the in vivo effects of betaarr2 on CXCR2-mediated neutrophil chemotaxis and on cutaneous wound healing. Surprisingly, the recruitment of neutrophils was increased in response to CXCL1 in the air pouch model and in the excisional wound beds of betaarr2(-/-) mice. Wound re-epithelialization was also significantly faster in betaarr2(-/-) mice than in betaarr2(+/+) mice. Taken together, the data indicate that betaarr2 is a negative regulator for CXCR2 in vivo signaling.


Arrestins/deficiency , Arrestins/genetics , Models, Immunological , Receptors, Interleukin-8B/physiology , Signal Transduction/genetics , Animals , Cell Movement/physiology , Epithelium/physiology , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Neutrophils/physiology , Wound Healing/physiology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , beta-Arrestin 2 , beta-Arrestins
11.
Bioorg Med Chem ; 10(4): 869-74, 2002 Apr.
Article En | MEDLINE | ID: mdl-11836092

In the course of our investigations of pyrimidines as antimycotic agents, we have identified a sub-class, with significant in vitro activity against mycobacteria. The salient feature of these pyrimidine derivatives (3a-o and 7a,b) is their appended aryl, heteroaryl and alkylthio substituent at position 6 and also alkylthio substituent at position 2. The rational design, synthesis, and evaluation of the in vitro antibacterial activity against six pathogenic bacteria including virulent and non-virulent strains of Mycobacterium tuberculosis is described. Some of the synthesized compounds (3c, 3h, 3i, 3o) have displayed only potent in vitro antimycobacterial activity with MIC of 0.75 microg/mL except 3i which also demonstrated activity against Escherichia coli at 12.5 microg/mL concentration. Only two compounds, 3a and 3b, demonstrated antibacterial activity against Pseudomonas aeruginosa and E. coli with MIC 12.5 microg/mL. All the synthesized compounds were also evaluated for their antimycotic activity against five pathogenic fungi but only some of them 3j-n and 7a,b were found most potent against Aspergillus fumigatus and Trichophyton mentagrophytes.


Anti-Infective Agents/chemical synthesis , Pyrimidines/pharmacology , Anti-Bacterial Agents , Anti-Infective Agents/pharmacology , Bacteria/drug effects , Chlorine , Drug Design , Fungi/drug effects , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Pyrimidines/chemical synthesis , Structure-Activity Relationship
...