Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 12: 28, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24739671

ABSTRACT

BACKGROUND: The Ron receptor tyrosine kinase (RTK) has been implicated in the progression of a number of carcinomas, thus understanding the regulatory mechanisms governing its activity is of potential therapeutic significance. A critical role for the juxtamembrane domain in regulating RTK activity is emerging, however the mechanism by which this regulation occurs varies considerably from receptor to receptor. RESULTS: Unlike other RTKs described to date, tyrosines in the juxtamembrane domain of Ron are inconsequential for receptor activation. Rather, we have identified an acidic region in the juxtamembrane domain of Ron that plays a central role in promoting receptor autoinhibition. Furthermore, our studies demonstrate that phosphorylation of Y1198 in the kinase domain promotes Ron activation, likely by relieving the inhibitory constraints imposed by the juxtamembrane domain. CONCLUSIONS: Taken together, our experimental data and molecular modeling provide a better understanding of the mechanisms governing Ron activation, which will lay the groundwork for the development of novel therapeutic approaches for targeting Ron in human malignancies.


Subject(s)
Receptor Protein-Tyrosine Kinases/chemistry , Amino Acid Sequence , HEK293 Cells , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/metabolism
2.
Crit Rev Immunol ; 33(6): 549-74, 2013.
Article in English | MEDLINE | ID: mdl-24266348

ABSTRACT

Numerous studies have documented abnormal expression and activation of the Ron receptor tyrosine kinase in a variety of human malignancies. Here we review the literature regarding the molecular mechanisms governing Ron regulation, the biological functions of Ron, the effect of Ron on cancer development, and potential therapeutic implications. In epithelial cells, activation of Ron by its ligand, macrophage stimulating protein, mediates a number of biological events including cell growth, motility, and epithelial to mesenchymal transition. Overexpression and/or activation of Ron has been implicated in the progression and metastasis of diverse epithelial cancers, where it plays a causal role in tumor development by promoting growth, survival, and motility of tumor cells. As a crucial regulator of inflammation, Ron inhibits classic macrophage activation and promotes alternative activation of macrophages, resulting in the resolution of inflammation and tissue repair. In addition, Ron alleviates antitumor immunity by promoting the alternative activation of tumor-associated macrophages, and Ron expression in the tumor microenvironment promotes the outgrowth of metastatic colonies. Hence, Ron is a promising therapeutic target for the treatment of epithelial cancers.


Subject(s)
Epithelial Cells/physiology , Inflammation/metabolism , Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Carcinogenesis , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Macrophage Activation , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/immunology
3.
J Immunol ; 187(5): 2181-92, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21810604

ABSTRACT

M1 activation of macrophages promotes inflammation and immunity to intracellular pathogens, whereas M2 macrophage activation promotes resolution of inflammation, wound healing, and tumor growth. These divergent phenotypes are characterized, in part, by the expression of inducible NO synthase and arginase I (Arg1) in M1 versus M2 activated macrophages, respectively. In this study, we demonstrate that the Ron receptor tyrosine kinase tips the balance of macrophage activation by attenuating the M1 phenotype while promoting expression of Arg1 through a Stat6-independent mechanism. Induction of the Arg1 promoter by Ron is mediated by an AP-1 site located 433 bp upstream of the transcription start site. Treatment of primary macrophages with macrophage stimulating protein, the ligand for Ron, induces potent MAPK activation, upregulates Fos, and enhances binding of Fos to the AP-1 site in the Arg1 promoter. In vivo, Arg1 expression in tumor-associated macrophages (TAMs) from Ron(-/-) mice was significantly reduced compared with that in TAMs from control animals. Furthermore, we show that Ron is expressed specifically by Tie2-expressing macrophages, a TAM subset that exhibits a markedly skewed M2 and protumoral phenotype. Decreased Arg1 in TAMs from Ron(-/-) mice was associated with reduced syngeneic tumor growth in these animals. These findings indicate that Ron induces Arg1 expression in macrophages through a previously uncharacterized AP-1 site in the Arg1 promoter and that Ron could be therapeutically targeted in the tumor microenvironment to inhibit tumor growth by targeting expression of Arg1.


Subject(s)
Arginase/biosynthesis , Gene Expression Regulation/immunology , Macrophages/enzymology , Neoplasms, Experimental/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factor AP-1/metabolism , Animals , Arginase/genetics , Arginase/immunology , Cell Separation , Flow Cytometry , Gene Expression , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology
4.
Am J Physiol Gastrointest Liver Physiol ; 300(3): G470-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193529

ABSTRACT

Dietary iron is particularly critical during periods of rapid growth such as in neonatal development. Human and rodent studies have indicated that iron deficiency or excess during this critical stage of development can have significant long- and short-term consequences. Since the requirement for iron changes during development, the availability of adequate iron is critical for the differentiation and maturation of individual organs participating in iron homeostasis. We have examined in rats the effects of dietary iron supplement following neonatal iron deficiency on tissue iron status in relation to erythropoietic ability during 16 wk of postweaning development. This physiological model indicates that postweaning iron-adequate diet following neonatal iron deficiency adversely affects erythroid differentiation in the bone marrow and promotes splenic erythropoiesis leading to splenomegaly and erythrocytosis. This altered physiology of iron homeostasis during postweaning development is also reflected in the inability to maintain liver and spleen iron concentrations and the altered expression of iron regulatory proteins in the liver. These studies provide critical insights into the consequences of neonatal iron deficiency and the dietary iron-induced cellular signals affecting iron homeostasis during early development.


Subject(s)
Anemia, Iron-Deficiency/blood , Bone Marrow/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis , Iron Deficiencies , Iron, Dietary/blood , Liver/metabolism , Spleen/metabolism , Age Factors , Anemia, Iron-Deficiency/diet therapy , Anemia, Iron-Deficiency/pathology , Animals , Animals, Newborn , Bone Marrow/pathology , Erythropoietin/blood , Female , Growth Differentiation Factor 15/blood , Hematocrit , Hemoglobins/metabolism , Homeostasis , Iron, Dietary/administration & dosage , Iron, Dietary/adverse effects , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/metabolism , Male , Maternal Nutritional Physiological Phenomena , Polycythemia/blood , Polycythemia/etiology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Spleen/pathology , Splenomegaly , Transferrin/metabolism , Weaning
5.
J Immunol ; 185(12): 7309-16, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21078906

ABSTRACT

The RON receptor tyrosine kinase regulates the balance between classical (M1) and alternative (M2) macrophage activation. In primary macrophages, the ligand for Ron, macrophage-stimulating protein (MSP), inhibits the expression of inducible NO synthase, a marker of classically activated macrophages, whereas promoting the expression of arginase I, a marker of alternative activation. Ron(-/-) mice express increased levels of IL-12, a product of classically activated macrophages, after endotoxin administration, resulting in increased serum IFN-γ levels and enhanced susceptibility to septic shock. In this study, we demonstrate that MSP inhibits LPS-induced IL-12p40 expression, and this inhibition is dependent on the docking site tyrosines in Ron. To further define this inhibition, we examined the effect of Ron on signaling pathways downstream of Ron. We found that MSP does not inhibit the MyD88-independent activation of IFN regulatory factor 3 and production of IFN-ß in response to LPS, nor does it inhibit MyD88-dependent TGF-ß-activated kinase phosphorylation or MAPK activation in primary macrophages. However, the induction of IκB kinase activity, IκB degradation, and DNA binding of NF-κB after LPS stimulation is delayed in the presence of MSP. In addition, Ron inhibits serine phosphorylation of p65 and NF-κB transcriptional activity induced by LPS stimulation of TLR4. Finally, MSP inhibits the NF-κB-dependent upregulation of the nuclear IκB family member, IκBζ, a positive regulator of secondary response genes including IL-12p40. LPS also induces expression of Ron and an N-terminally truncated form of Ron, Sf-Ron, in primary macrophages, suggesting that the upregulation of Ron by LPS could provide classical feedback regulation of TLR signaling.


Subject(s)
Hepatocyte Growth Factor/immunology , I-kappa B Kinase/immunology , Macrophages/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Cell Line , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Protein Binding/genetics , Protein Binding/immunology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/genetics , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription Factor RelA/metabolism
6.
J Leukoc Biol ; 88(2): 373-82, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20453108

ABSTRACT

Acute endotoxemia is associated with excessive production of proinflammatory mediators by hepatic macrophages and endothelial cells, which have been implicated in liver injury and sepsis. In these studies, we analyzed the role of MSP and its receptor STK in regulating the activity of these cells. Acute endotoxemia, induced by administration of LPS (3 mg/kg) to mice, resulted in increased expression of STK mRNA and protein in liver macrophages and endothelial cells, an effect that was dependent on TLR-4. This was correlated with decreased MSP and increased pro-MSP in serum. In Kupffer cells, but not endothelial cells, MSP suppressed LPS-induced NOS-2 expression, with no effect on COX-2. LPS treatment of mice caused a rapid (within 3 h) increase in the proinflammatory proteins NOS-2, IL-1beta, and TNF-alpha, as well as TREM-1 and TREM-3 and the anti-inflammatory cytokine IL-10 in liver macrophages and endothelial cells. Whereas LPS-induced expression of proinflammatory proteins was unchanged in STK-/- mice, IL-10 expression was reduced significantly. Enzymes mediating eicosanoid biosynthesis including COX-2 and mPGES-1 also increased in macrophages and endothelial cells after LPS administration. In STK-/- mice treated with LPS, mPGES-1 expression increased, although COX-2 expression was reduced. LPS-induced up-regulation of SOD was also reduced in STK-/- mice in liver macrophages and endothelial cells. These data suggest that MSP/STK signaling plays a role in up-regulating macrophage and endothelial cell anti-inflammatory activity during hepatic inflammatory responses. This may be important in protecting the liver from tissue injury.


Subject(s)
Endotoxemia/immunology , Liver/pathology , fms-Like Tyrosine Kinase 3/physiology , Acute Disease , Animals , Endothelial Cells/metabolism , Inflammation/immunology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/pharmacology , Liver/immunology , Liver Diseases/immunology , Liver Diseases/pathology , Macrophages/metabolism , Mice , Mice, Knockout , RNA, Messenger/drug effects , Serine Endopeptidases/physiology , Signal Transduction/immunology , fms-Like Tyrosine Kinase 3/genetics
7.
J Virol ; 84(5): 2223-35, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20016000

ABSTRACT

Friend virus induces an erythroleukemia in susceptible mice that is initiated by the interaction of the Friend virus-encoded glycoprotein gp55 with the erythropoietin (Epo) receptor and the product of the host Fv2 gene, a naturally occurring truncated form of the Stk receptor tyrosine kinase (Sf-Stk). We have previously demonstrated that the activation of Sf-Stk, recruitment of a Grb2/Gab2/Stat3 signaling complex, and induction of Pu.1 expression by Stat3 are required for the development of the early stage of Friend disease both in vitro and in vivo. Here we demonstrate that the interaction of gp55 with Sf-Stk is dependent on cysteine residues in the ecotropic domain of gp55 and the extracellular domain of Sf-Stk. Point mutation of these cysteine residues or deletion of these domains inhibits the ability of gp55 to interact with Sf-Stk, resulting in the inability of these proteins to promote the Epo-independent growth of erythroid progenitor cells. We also demonstrate that the interaction of gp55 with Sf-Stk does not promote dimerization of Sf-Stk but results in enhanced phosphorylation of Sf-Stk and the relocalization of Sf-Stk from the cytosol to the plasma membrane. Finally, we demonstrate that a constitutively active form of Sf-Stk (Sf-StkM330T), as well as its human counterpart, Sf-Ron, promotes Epo-independent colony formation in the absence of gp55 and that this response is also dependent on the cysteines in the extracellular domains of Sf-StkM330T and Sf-Ron. These data suggest that the cysteines in the extracellular domains of Sf-Stk and Sf-Ron may also mediate the interaction of these truncated receptors with other cellular factors that regulate their ability to promote cytokine-independent growth.


Subject(s)
Cysteine/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Viral Envelope Proteins/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Cysteine/genetics , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoietin/genetics , Erythropoietin/metabolism , Humans , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Protein Conformation , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
8.
Front Biosci (Landmark Ed) ; 14(14): 5273-90, 2009 06 01.
Article in English | MEDLINE | ID: mdl-19482615

ABSTRACT

Activation of the Jak/Stat pathway plays a central role in hematopoietic development. Here, we focus on unique functions for Stat3 in the development of the hematopoietic system. Recent studies highlight a critical role for Stat3 in the development of T helper cell and B cell subsets as well as dendritic cell development and maturation. Further, Stat3 appears to play an important function in limiting neutrophil numbers and in regulating hematopoietic stem cell self-renewal. In macrophages, Stat3 is essential in limiting TLR signaling and the inflammatory damage associated with classical macrophage activation. Stat3 also plays an essential role in leukemic development induced by viral oncogenes, chromosomal translocations and by the Friend erythroleukemia virus. We also discuss the mechanism by which Stat3 is activated by extracellular signals, and the current understanding of the role of post-translational modification in the regulation of Stat3 activity. Understanding the molecular events underlying Stat3 activation will be critical in generating targeted therapeutics aimed at modulating Stat3 function.


Subject(s)
Hematopoiesis/physiology , STAT3 Transcription Factor/physiology , Animals , Cell Differentiation , Cell Transformation, Neoplastic , Humans , Immunity, Innate , Macrophage Activation , Models, Biological , Protein Processing, Post-Translational , Receptors, Cytokine/physiology , Receptors, Growth Factor/physiology , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology
9.
J Immunol ; 181(4): 2303-10, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684919

ABSTRACT

Receptor tyrosine kinases are emerging as a class of key regulators of innate immune responses. We have shown previously that the RON receptor tyrosine kinases (murine Stk), expressed on tissue-resident macrophages, inhibit classical macrophage activation while promoting hallmarks of alternative activation, thus regulating the critical balance between the inflammatory and wound-healing properties of activated macrophages. We have also shown previously that RON(-/-) mice are more susceptible to in vivo endotoxin challenge than wild-type mice, suggesting that the expression of this receptor confers a degree of endotoxin resistance to these animals. Here we demonstrate that, in response to in vivo LPS challenge, RON(-/-) mice harbor significantly increased systemic levels of IFN-gamma and IL-12p70 and increased levels of IL-12p40 transcript in their spleen. This elevation of IFN-gamma can be attributed to splenic NK cells responding to the elevated levels of IL-12. Analysis of RON and IFN-gamma receptor double-knockout mice indicates that the enhanced susceptibility of RON(-/-) mice to endotoxin challenge is dependent on IFN-gamma-mediated signals. In vitro studies demonstrate that stimulation of primary peritoneal macrophages with macrophage-stimulating protein, the ligand for RON, inhibits IFN-gamma-induced STAT1 phosphorylation and CIITA expression, resulting in reduced surface levels of MHC class II. Further studies demonstrating the induction of suppressor of cytokine signaling 1 via macrophage-stimulating protein/RON signaling provide a potential mechanistic insight into this regulatory pathway. These results indicate that the RON receptor regulates both the production of and response to IFN-gamma, resulting in enhanced susceptibility to endotoxin challenge.


Subject(s)
Immunity, Innate , Interferon-gamma/biosynthesis , Receptor Protein-Tyrosine Kinases/physiology , Up-Regulation/immunology , Animals , Cells, Cultured , Genetic Predisposition to Disease , Immunity, Innate/genetics , Interferon-gamma/blood , Interferon-gamma/physiology , Interleukin-12/biosynthesis , Interleukin-12/blood , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Subunits/biosynthesis , Protein Subunits/blood , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Interferon/physiology , Shock, Septic/genetics , Shock, Septic/immunology , Signal Transduction/immunology , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Interferon gamma Receptor
10.
J Immunol ; 181(2): 1548-55, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606710

ABSTRACT

HIV encodes several proteins, including Tat, that have been demonstrated to modulate the expression of receptors critical for innate immunity, including MHC class I, mannose receptor, and beta(2)-microglobulin. We demonstrate that Tat targets the receptor tyrosine kinase recepteur d'origine nantais (RON), which negatively regulates inflammation and HIV transcription, for proteosome degradation. Tat decreases cell surface RON expression in HIV-infected monocytic cells, and Tat-mediated degradation of RON protein is blocked by inhibitors of proteosome activity. Tat specifically induced down-regulation of RON and not other cell surface receptors, such as the transferrin receptor, the receptor tyrosine kinase TrkA, or monocytic markers CD14 and ICAM-1. The Tat trans activation domain is required for RON degradation, and this down-regulation is dependent on the integrity of the kinase domain of RON receptor. We propose that Tat mediates degradation of RON through a ubiquitin-proteosome pathway, and suggest that by targeting signals that modulate inflammation, Tat creates a microenvironment that is optimal for HIV replication and progression of AIDS-associated diseases.


Subject(s)
HIV-1 , Macrophages/virology , Receptor Protein-Tyrosine Kinases/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Genetic Vectors , HIV-1/immunology , Humans , Inflammation , Macrophages/immunology , Macrophages/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/chemistry , Transfection , U937 Cells , Ubiquitin/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemistry
11.
J Immunol ; 180(3): 1670-7, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209063

ABSTRACT

Efficient HIV-1 transcription requires the induction of cellular transcription factors, such as NF-kappaB, and the viral factor Tat, which through the recruitment of P-TEFb enhances processive transcription. However, whether cellular signals repress HIV-1 transcription to establish proviral latency has not been well studied. Previously, it has been shown that the receptor tyrosine kinase RON inhibits HIV transcription. To gain insights into the biochemical mechanisms by which RON inhibits transcription we examined the binding of transcription factors to the HIV provirus long terminal repeat using chromatin immunoprecipitation. RON expression decreased basal levels of NF-kappaB and RNA polymerase II (Pol II) binding to the HIV provirus long terminal repeat but did not prevent the induction of these complexes following treatment with cytokines. However, RON did decrease efficient transcription elongation because reduced RNA Pol II was associated with HIV-1 genomic sequences downstream of the transcriptional start site. There was a correlation between RON expression and increased binding of factors that negatively regulate transcription elongation, NELF, Spt5, and Pcf11. Furthermore, the ability of RON to inhibit HIV-1 transcription was sensitive to a histone deacetylase inhibitor and was associated with nucleosome remodeling. These results indicate that RON represses HIV transcription at multiple transcriptional check points including initiation, elongation and chromatin organization and are the first studies to show that cellular signaling pathways target Pol II pausing to repress gene expression.


Subject(s)
Gene Expression Regulation, Viral , HIV-1/genetics , RNA Polymerase II/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Cell Line , Chromatin Immunoprecipitation , HIV Long Terminal Repeat , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nucleosomes/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...