Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 9(12): e116141, 2014.
Article in English | MEDLINE | ID: mdl-25551463

ABSTRACT

Eosinophils are produced in the bone marrow from CD34+ eosinophil lineage-committed progenitors, whose levels in the bone marrow are elevated in a variety of human diseases. These findings suggest that increased eosinophil lineage-committed progenitor production is an important process in disease-associated eosinophilia. The pathways central to the biology of the eosinophil lineage-committed progenitor remain largely unknown. Thus, developing new methods to investigate the regulators of eosinophil lineage-committed progenitor differentiation is needed to identify potential therapeutic targets to specifically inhibit eosinophil production. We tested cytokine regimens to optimize liquid cultures for the study of eosinophil lineage-committed progenitor and eosinophil precursor differentiation into mature eosinophils. Stem cell factor (but not fms-related tyrosine kinase 3 ligand) was required for optimal yield of eosinophils. Furthermore, we evaluated the effects of cell preservation and scale on the culture, successfully culturing functional eosinophils from fresh and frozen murine bone marrow cells and in a standard-sized and 96-well culture format. In summary, we have developed an adaptable culture system that yields functionally competent eosinophils from murine low-density bone marrow cells and whose cytokine regime includes expansion of progenitors with stem cell factor alone with subsequent differentiation with interleukin 5.


Subject(s)
Bone Marrow Cells/cytology , Eosinophils/cytology , Stem Cells/cytology , Animals , Antigens, CD34/metabolism , Cell Culture Techniques , Cells, Cultured , Cryopreservation , Female , Hematopoiesis , Leukocyte Count , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology
2.
Cancer Res ; 73(6): 1752-63, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23328584

ABSTRACT

Ron receptor kinase (MST1R) is important in promoting epithelial tumorigenesis, but the potential contributions of its specific expression in stromal cells have not been examined. Herein, we show that the Ron receptor is expressed in mouse and human stromal cells of the prostate tumor microenvironment. To test the significance of stromal Ron expression, prostate cancer cells were orthotopically implanted into the prostates of either wild-type or Ron tyrosine kinase deficient (TK(-/-); Mst1r(-/-)) hosts. In TK(-/-) hosts, prostate cancer cell growth was significantly reduced as compared with tumor growth in TK(+/+) hosts. Prostate tumors in TK(-/-) hosts exhibited an increase in tumor cell apoptosis, macrophage infiltration and altered cytokine expression. Reciprocal bone marrow transplantation studies and myeloid cell-specific ablation of Ron showed that loss of Ron in myeloid cells is sufficient to inhibit prostate cancer cell growth. Interestingly, depletion of CD8(+) T cells, but not CD4(+) T cells, was able to restore prostate tumor growth in hosts devoid of myeloid-specific Ron expression. These studies show a critical role for the Ron receptor in the tumor microenvironment, whereby Ron loss in tumor-associated macrophages inhibits prostate cancer cell growth, at least in part, by derepressing the activity of CD8(+) T cells.


Subject(s)
Prostatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Flow Cytometry , Immunohistochemistry , Male , Mice , Mice, Knockout , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/immunology , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics
3.
FASEB J ; 26(10): 4092-101, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22787265

ABSTRACT

To systematically identify novel gene functions essential for osteogenesis and skeletal mineralization, we performed a forward genetic mutagenesis screen in zebrafish and isolated a mutant that showed delayed skeletal mineralization. Analysis of the mutant phenotype in an osterix:nuclear-GFP transgenic background demonstrated that mutants contain osterix-expressing osteoblasts comparable to wild-type embryos. Positional cloning revealed a premature stop mutation in the macrophage-stimulating protein (msp) gene, predicted to result in a biologically inactive protein. Analysis of the embryonic expression pattern for the receptor for Msp, Ron, shows specific expression in the corpuscles of Stannius, a teleost-specific organ that produces stanniocalcin, a pivotal hormone in fish calcium homeostasis. Knockdown of Ron resulted in identical phenotypes as observed in msp mutants. Msp mutant embryos could be rescued by excess calcium. Consistent with a role for Msp/Ron in calcium homeostasis, calcium-regulating factors, such as pth1, pth2, stc1l, and trpv5/6 were significantly affected in msp mutant larvae. While Msp and Ron have previously been shown to play a critical role in a wide variety of biological processes, we introduce here the Msp/Ron signaling axis as a previously unappreciated player in calcium homeostasis and embryonic skeletal mineralization.


Subject(s)
Calcium/metabolism , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Glycoproteins/metabolism , Hepatocyte Growth Factor/genetics , Homeostasis/genetics , Homeostasis/physiology , Osteogenesis/genetics , Osteogenesis/physiology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics
4.
Endocrinology ; 153(6): 2735-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22474186

ABSTRACT

The Ron receptor tyrosine kinase (macrophage stimulating 1 receptor) is overexpressed in approximately 50% of human breast cancers. Transgenic mice overexpressing Ron in the mammary epithelium [mouse mammary tumor virus driven (MMTV)-Ron expressing mice] develop mammary tumors that exhibit up-regulation of ß-catenin and ß-catenin target genes. ß-Catenin has been shown to be a mediator of mammary tumorigenesis in various breast cancer models, including downstream of Ron. However, the in vivo impact of a conditional loss of ß-catenin downstream of Ron receptor overexpression on the onset, growth, turnover, and metastasis of mammary tumors has not been addressed. To determine the significance of ß-catenin in the context of Ron overexpression, we conditionally deleted ß-catenin in mammary epithelial cells of MMTV-Ron mice. Conditional deletion of ß-catenin in the mammary epithelium, through the use of whey acidic protein (WAP)-Cre transgenic mice, significantly delayed the onset of mammary hyperplastic nodules, the presence of palpable mammary tumors, and ultimately decreased liver metastasis. ß-Catenin loss in this model was also associated with decreased expression of cyclin D1. In total, these studies support an important role for ß-catenin downstream of Ron receptor signaling during the development of mammary tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mammary Glands, Animal/metabolism , Receptor Protein-Tyrosine Kinases/genetics , beta Catenin/genetics , Animals , Blotting, Western , Cyclin D1/genetics , Cyclin D1/metabolism , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Hyperplasia , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Mice, Transgenic , Receptor Protein-Tyrosine Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , beta Catenin/deficiency
5.
Mol Cancer ; 11: 2, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22226043

ABSTRACT

BACKGROUND: The receptor tyrosine kinase family includes many transmembrane proteins with diverse physiological and pathophysiological functions. The involvement of tyrosine kinase signaling in promoting a more aggressive tumor phenotype within the context of chemotherapeutic evasion is gaining recognition. The Ron receptor is a tyrosine kinase receptor that has been implicated in the progression of breast cancer and evasion of tamoxifen therapy. RESULTS: Here, we report that Ron expression is correlated with in situ, estrogen receptor alpha (ERα)-positive tumors, and is higher in breast tumors following neoadjuvant tamoxifen therapy. We also demonstrate that the majority of mammary tumors isolated from transgenic mice with mammary specific-Ron overexpression (MMTV-Ron mice), exhibit appreciable ER expression. Moreover, genetic-ablation of ERα, in the context of Ron overexpression, leads to delayed mammary tumor initiation and growth, but also results in an increased metastasis. CONCLUSIONS: Ron receptor overexpression is associated with ERα-positive human and murine breast tumors. In addition, loss of ERα on a Ron overexpressing background in mice leads to the development of breast tumors which grow slower but which exhibit more metastasis and suggests that targeting of ERα, as in the case of tamoxifen therapy, may reduce the growth of Ron overexpressing breast cancers but may cause these tumors to be more metastatic.


Subject(s)
Estrogen Receptor alpha/genetics , Gene Deletion , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Receptor Protein-Tyrosine Kinases/genetics , Animals , Cell Proliferation , Estrogen Receptor alpha/metabolism , Female , Gene Expression , Mammary Neoplasms, Animal/mortality , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Staging , Phenotype , Receptor Protein-Tyrosine Kinases/metabolism
6.
Cancer Lett ; 314(1): 92-101, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22004727

ABSTRACT

Previous studies have shown that the Ron receptor is overexpressed in prostate cancer and Ron expression increases with disease severity in humans and the mouse TRAMP model. Here, the causal role of Ron overexpression in the murine prostate was examined in the development and progression of prostate cancer. Transgenic mouse strains were generated which selectively overexpressed Ron in the prostate epithelium and prostate histopathology was evaluated and compared to wild type controls. Ron overexpression led to the development of prostate intraepithelial neoplasia (mPIN) with local invasion and was associated with increases in prostate cell proliferation and decreases in cell death.


Subject(s)
Prostatic Intraepithelial Neoplasia/etiology , Prostatic Neoplasms/etiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Apoptosis , Cell Proliferation , In Situ Nick-End Labeling , Male , Mice , Mice, Transgenic , Prostate/chemistry , Prostate/pathology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/analysis , Signal Transduction
7.
Hepatology ; 53(5): 1618-28, 2011 May.
Article in English | MEDLINE | ID: mdl-21520175

ABSTRACT

UNLABELLED: Previous studies demonstrated that targeted deletion of the Ron receptor tyrosine kinase (TK) domain in mice leads to marked hepatocyte protection in a well-characterized model of lipopolysaccharide (LPS)-induced acute liver failure in D-galactosamine (GalN)-sensitized mice. Hepatocyte protection in TK-/- mice was observed despite paradoxically elevated serum levels of tumor necrosis factor alpha (TNF-α). To understand the role of Ron in the liver, purified populations of Kupffer cells and hepatocytes from wildtype (TK+/+) and TK-/- mice were studied. Utilizing quantitative reverse-transcription polymerase chain reaction (RT-PCR), we demonstrated that Ron is expressed in these cell types. Moreover, we also recapitulated the protected hepatocyte phenotype and exaggerated cytokine production observed in the TK-/- mice in vivo through the use of purified cultured cells ex vivo. We show that isolated TK-/- Kupffer cells produce increased levels of TNF-α and select cytokines compared to TK+/+ cells following LPS stimulation. We also show that conditioned media from LPS-treated TK-/- Kupffer cells was more toxic to hepatocytes than control media, suggesting the exaggerated levels of cytokines produced from the TK-/- Kupffer cells are detrimental to wildtype hepatocytes. In addition, we observed that TK-/- hepatocytes were more resistant to cell death compared to TK+/+ hepatocytes, suggesting that Ron functions in both the epithelial and inflammatory cell compartments to regulate acute liver injury. These findings were confirmed in vivo in mice with hepatocyte and macrophage cell-type-specific conditional Ron deletions. Mice with Ron loss selectively in hepatocytes exhibited less liver damage and increased survival compared to mice with Ron loss in macrophages. CONCLUSION: We dissected cell-type-specific roles for Ron such that this receptor modulates cytokine production from Kupffer cells and inhibits hepatocyte survival in response to injury.


Subject(s)
Hepatocytes/drug effects , Kupffer Cells/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Cell Survival/drug effects , Cells, Cultured , Endotoxins/pharmacology , Hepatocytes/physiology , Male , Mice , Mice, Inbred C57BL
8.
Mol Cancer Ther ; 10(1): 29-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21220489

ABSTRACT

The proliferation cell nuclear antigen (PCNA) is a critical protein required for DNA replication in proliferating cells including cancer cells. However, direct inhibition of PCNA in cancer cells has been difficult due to the lack of targetable sites. We previously reported that phosphorylation of tyrosine 211 (Y211) on PCNA is important for the proliferative function of PCNA when this protein is associated with the chromatin in cancer cells. Here, we show that the Y211 phosphorylation of PCNA is a frequent event in advanced prostate cancer. To explore the potential of this signaling event in inhibition of cancer cell growth, we used a synthetic peptide, the Y211F peptide, which when present inhibits phosphorylation of Y211 on endogenous PCNA. Treatment with this peptide, but not a scrambled control peptide, resulted in S-phase arrest, inhibition of DNA synthesis, and enhanced cell death in a panel of human prostate cancer cell lines. In addition, treatment with the Y211F peptide led to decreased tumor growth in PC3-derived xenograft tumors in vivo in nude mice. Our study shows for the first time that PCNA phosphorylation at Y211 is a frequent and biologically important signaling event in prostate cancer. This study also shows a proof of concept that Y211 phosphorylation of PCNA may be used as a therapeutic target in prostate cancer cells, including cells of advanced cancers that are refractory to standard hormonal therapies.


Subject(s)
Peptides/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Amino Acid Sequence , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Molecular Sequence Data , Molecular Targeted Therapy , Phosphorylation/drug effects , Proliferating Cell Nuclear Antigen/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tyrosine/metabolism , Xenograft Model Antitumor Assays
9.
Innate Immun ; 17(6): 499-507, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21088048

ABSTRACT

Previous studies have shown that the Ron receptor tyrosine kinase is an important regulator of the acute lung inflammatory response induced by intranasal administration of bacterial LPS. Compared to wild-type mice, complete loss of the Ron receptor in all cell types in vivo was associated with increased lung damage as determined by histological analyses and several markers of lung injury including increases in pro-inflammatory cytokines such as TNF-α. Tumor-necrosis factor-α is a multifunctional cytokine secreted by macrophages, which plays a major role in inflammation and is a central mediator of several disease states including rheumatoid arthritis and sepsis. Based on increased TNF-α production observed in the Ron-deficient mice, we hypothesized that Ron receptor function in the inflammatory cell compartment is essential for the regulating lung injury in vivo. To test this hypothesis, we generated myeloid lineage-specific Ron-deficient mice. In this study, we report that loss of Ron signaling selectively in myeloid cells results in increased lung injury following intranasal administration of LPS as measured by increases in TNF-α production, ensuing neutrophil accumulation and increased lung histopathology. These findings corroborate the role of Ron receptor tyrosine kinase as a negative regulator of inflammation and further demonstrate the in vivo significance of Ron signaling selectively in myeloid cells as a major regulator of this response in vivo. These data authenticate Ron as a potential target in innate immunity and TNF-α-mediated pathologies.


Subject(s)
Acute Lung Injury/pathology , Myeloid Cells/pathology , Pulmonary Alveoli/pathology , Receptor Protein-Tyrosine Kinases/deficiency , Tumor Necrosis Factor-alpha/biosynthesis , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Gene Expression , Immunity, Innate/drug effects , Immunity, Innate/physiology , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/genetics
10.
Shock ; 33(2): 197-204, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19487969

ABSTRACT

The Ron receptor tyrosine kinase (TK) plays a regulatory role in the inflammatory response to acute lung injury induced by intranasal administration of bacterial LPS. Previously, we have shown that mice with a targeted deletion of the TK signaling domain of the Ron receptor exhibited more severe lung injury in response to intranasal LPS administration as evidenced by an increased leakage of albumin in the lungs and a greater thickening of the alveolar septa compared with wild-type mice. In addition, lung injury in the Ron TK-deficient (TK(-/-)) mice was associated with increased activation of the transcription factor, nuclear factor-kappaB (NF-kappaB), and significantly increased intrapulmonary expression of TNFalpha. TNFalpha, a multifunctional proinflammatory cytokine, is a central mediator in several disease states, including rheumatoid arthritis and sepsis. On the basis of the observation that TNFalpha production is increased in the Ron TK-/- mice and that macrophages are a major source of this cytokine, we hypothesized that the alterations observed in the Ron TK(-/-) mice may be due, in part, to Ron signaling, specifically in alveolar macrophages. To test this hypothesis, we used the wild-type and Ron TK(-/-) primary alveolar macrophages and the murine alveolar macrophage cell line, MH-S, to examine the effects of Ron activation on LPS-induced TNFalpha production and NF-kappaB activity. Here, we reported that Ron is expressed on alveolar macrophages and MH-S cells. Activation of Ron by its ligand, hepatocyte growth factor-like protein, decreases TNFalpha production in alveolar macrophages after LPS challenge. Decreased TNFalpha is associated with hepatocyte growth factor-like protein-induced decreases in NF-kappaB activation and increases in the NF-kappaB inhibitory protein, IkappaB. We also provided the first evidence for Ron as a negative regulator of Adam17, the metalloprotease involved in TNFalpha processing. These results indicate that Ron plays a critical role in regulation of alveolar macrophage signaling and validates this receptor as a target in TNFalpha-mediated pulmonary pathologies.


Subject(s)
ADAM Proteins/metabolism , Macrophages, Alveolar/metabolism , NF-kappa B/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Tumor Necrosis Factor-alpha/metabolism , ADAM17 Protein , Animals , Cell Line , Cells, Cultured , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Male , Mice , Mice, Mutant Strains , Mutation , Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...