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











Publication year range
1.
Int J Cancer ; 139(10): 2270-6, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27459381

ABSTRACT

Sunitinib and sorafenib are broad-spectrum tyrosine kinase inhibitors (TKI) targeting, for example, VEGF1-3, PDGFRb, RET, FLT3, CD117 (c-KIT) and CSF-1R cell membrane receptors thus suppressing tumor angiogenesis and cancer cell growth. Recently it has been suggested that the kinases targeted by Sunitinib and/or Sorafenib regulate leukocyte transmigration, which might in part be responsible for the often-observed reduction in tumor-associated myeloid derived suppressor cells and regulatory T cells. The aim of the current study is to determine whether sunitinib or sorafenib inhibit leukocyte extravasation. Sunitinib, sorafenib, or vehicle treated animals did not show any difference in leukocyte trafficking either in peritonitis or in vivo homing experiments, although sunitinib treatment effectively inhibited growth of B16 melanoma tumors in WT, SCID and SCID beige mice. Inhibition of tumor growth was associated with an increased number of infiltrating CD11b+ cells in the tumor, while the numbers of CD8, Gr-1 and F4/80 expressing cells were unchanged. In conclusion, the findings suggest that despite multiple targets with a potential role in leukocyte extravasation, neither sunitinib nor sorafenib effectively inhibits this process in vivo. Thus, the observed specific effect on CD11b cells among tumor infiltrating leukocytes is most likely an indirect effect.


Subject(s)
Indoles/pharmacology , Leukocytes/drug effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Cell Movement/drug effects , Female , Leukocytes/enzymology , Leukocytes/immunology , Leukocytes/pathology , Melanoma, Experimental/blood , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Niacinamide/pharmacology , Sorafenib , Sunitinib , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transendothelial and Transepithelial Migration/drug effects
2.
PLoS One ; 10(8): e0134721, 2015.
Article in English | MEDLINE | ID: mdl-26258883

ABSTRACT

The ectoenzyme CD73 catalyzes the hydrolysis of AMP, and is one of the most important producers of extracellular adenosine. On regulatory T cells, CD73 is necessary for immunosuppressive functions, and on Th17 cells CD73-generated adenosine exerts anti-inflammatory effects. However, the expression and function of CD73 in pro-inflammatory M1 and in immunosuppressive M2 macrophages is largely unknown. Here we show that CD73 expression and enzyme activity were induced in in vitro polarized pro-inflammatory human M(LPS+TNF) monocytes/macrophages, while CD73 was absent from immunosuppressive M(IL-4+M-CSF)-polarized macrophages. Inhibition of CD73 activity with the inhibitor AMPCP did not affect the polarization of human monocytes. In mice, CD73 was present on resident peritoneal macrophages. In striking contrast, elicited peritoneal macrophages remained CD73 negative regardless of their polarization towards either a pro-inflammatory M(LPS) or anti-inflammatory M(IL-4c) direction. Finally, the ability of peritoneal macrophages to polarize to pro- and anti-inflammatory cells was perfectly normal in CD73-deficient mice in vivo. These data indicate that, in contrast to other major leukocyte subpopulations, CD73 activity on macrophages does not play a major role in their polarization and that in mice host CD73 on any cell type is not required in vivo for peritoneal macrophage polarization towards either a pro- or an anti-inflammatory direction.


Subject(s)
5'-Nucleotidase/metabolism , Macrophages, Peritoneal/metabolism , Adenosine/chemistry , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/chemistry , Animals , Cytokines/metabolism , Flow Cytometry , GPI-Linked Proteins/metabolism , Humans , Immunosuppressive Agents/chemistry , Inflammation , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lipopolysaccharides/chemistry , Macrophage Colony-Stimulating Factor/metabolism , Macrophages, Peritoneal/cytology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Phenotype
3.
Antioxid Redox Signal ; 21(10): 1460-74, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24328532

ABSTRACT

AIMS: Rat sarcoma virus (RAS)-induced tumorigenesis has been suggested to follow a three-stage model consisting of an initial RAS activation, senescence induction, and evasion of p53-dependent senescence checkpoints. While reactive oxygen species act as second messengers in RAS-induced senescence, they are also involved in oncogenic transformation by inducing proliferation and promoting mutations. In the current work, we investigated the role of extracellular superoxide dismutase (SOD3) in RAS-induced senescence and immortalization in vitro and in vivo. We used a mouse embryonic fibroblast (MEF) primary cell model along with immortalized and transformed human cell lines derived from papillary and anaplastic thyroid cancer. RESULTS: Based on our data, sod3 RNA interference in H-RasV12-transduced cells markedly inhibited cell growth, while sod3 over-expression in MEFs initially caused a proliferative burst followed by the activation of DNA damage checkpoints, induction of p53-p21 signal transduction, and senescence. Subsequently, sod3-transduced MEF cells developed co-operative p21-p16 down-regulation and acquired transformed cell characteristics such as increased telomerase activity, loss of contact inhibition, growth in low-nutrient conditions, and in vivo tumorigenesis. Interestingly, as previously reported with RAS, we showed a dose-dependent response to SOD3 in vitro and in vivo involving transcriptional and non-transcriptional regulatory mechanisms. INNOVATION: SOD3 may mediate H-RasV12-induced initiation of primary cell immortalization. CONCLUSIONS: Our results indicate that SOD3 influences growth signaling in primary and cancer cells downstream of the ras oncogene and could serve as a therapy target at an early tumorigenesis phase.


Subject(s)
Cell Transformation, Neoplastic , Embryo, Mammalian/metabolism , Superoxide Dismutase/metabolism , Animals , Embryo, Mammalian/cytology , Fibroblasts/cytology , Mice
4.
Stem Cells ; 31(6): 1218-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23404893

ABSTRACT

Mesenchymal stromal cells (MSCs) are able to influence the growth abilities of transformed cells. Here, we show that papillary thyroid cancer TPC1 and HEK 293T cells interact physically with human primary bone marrow-derived MSCs followed by evanescence of MSC cytoplasm. Interestingly, transformed cells were able to connect only to apoptotic MSCs that had lost their migration ability, whereas naïve MSCs avoided the direct contact. The interaction stimulated the proliferation of the cocultured transformed cells, activated mitogen and stress signaling, and increased resistance to cytotoxins. Consistent with in vitro data, the MSC interaction stimulated transformed cells had enhanced ability to grow and metastasize in vivo. The parental control cells showed mild tumorigenicity as compared to MSC interaction stimulated cells yielding measurable tumors in 31 days and 7 days, respectively. Our coculture model system describes how adjacent transformed cells absorb stromal cells thereby leading to the stroma-driven evolution of moderately carcinogenic cells to highly aggressive metastatic cells.


Subject(s)
Mesenchymal Stem Cells/pathology , Animals , Atrophy , Bone Marrow Cells/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Cells, Cultured , Coculture Techniques , HEK293 Cells , Humans , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Stromal Cells/pathology , Thyroid Neoplasms/pathology
5.
J Virol ; 86(24): 13779-84, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035221

ABSTRACT

Canine parvovirus (CPV) infection leads to reorganization of nuclear proteinaceous subcompartments. Our studies showed that virus infection causes a time-dependent increase in the amount of viral nonstructural protein NS1 mRNA. Fluorescence recovery after photobleaching showed that the recovery kinetics of nuclear transcription-associated proteins, TATA binding protein (TBP), transcription factor IIB (TFIIB), and poly(A) binding protein nuclear 1 (PABPN1) were different in infected and noninfected cells, pointing to virus-induced alterations in binding dynamics of these proteins.


Subject(s)
Parvoviridae Infections/metabolism , Subcellular Fractions/metabolism , Transcription Factors/metabolism , Animals , Cell Compartmentation , Parvovirus, Canine/isolation & purification
6.
PLoS One ; 6(8): e24456, 2011.
Article in English | MEDLINE | ID: mdl-21909393

ABSTRACT

BACKGROUND: Extracellular superoxide dismutase (SOD3), which dismutates superoxide anion to hydrogen peroxide, has been shown to reduce the free radical stress derived apoptosis in tissue injuries. Since both superoxide anion and hydrogen peroxide have a marked impact on signal transduction pathways and could potentially explain a number of apoptosis and survival -related phenomena in different pathological conditions, we clarified the impact of SOD3 on Akt and Erk1/2 cell survival pathways in rat hind limb injury model. METHODOLOGY AND PRINCIPAL FINDINGS: Based on our data, the hind limb ischemic rats treated with virally delivered sod3 have milder injury and less apoptosis than control animals that could be due to parallel activation of pro-proliferative and anti-apoptotic Erk1/2 and Akt pathways. The common downstream factor of both signaling pathways, the apoptosis related forkhead box protein O3a (FoxO3a), was phosphorylated and translocated to the cytoplasm in sod3 treated tissues and cell line. Additionally, we obtained increased mRNA production of elk-1, ets-1, and microRNA 21 (miR-21), whereas synthesis of bim mRNA was decreased in sod3 overexpressing tissues. We further showed that overexpression of sod3 modulated redox related gene expression by downregulating nox2 and inos when compared to injured control animals. CONCLUSIONS AND SIGNIFICANCE: The study shows the complexity of SOD3-derived effects on tissue injury recovery that are not limited to the reduction of superoxide anion caused cellular stress but highlights the impact of SOD3 related signal transduction on tissue functions and suggests an important role for SOD3 in attenuating cell stress effects in different pathological conditions.


Subject(s)
Apoptosis , Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Transcription Factors/metabolism , Ischemia/enzymology , Ischemia/pathology , Proto-Oncogene Proteins c-akt/metabolism , Superoxide Dismutase/metabolism , Animals , Cell Survival , Forkhead Box Protein O3 , MAP Kinase Signaling System , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , NIH 3T3 Cells , Phosphorylation , Rats , Rats, Inbred F344
7.
Eur J Immunol ; 41(5): 1231-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21469131

ABSTRACT

CD73/ecto-5'-nucleotidase dephosphorylates extracellular AMP into adenosine, and it is a key enzyme in the regulation of adenosinergic signaling. The contribution of host CD73 to tumor growth and anti-tumor immunity has not been studied. Here, we show that under physiological conditions CD73-deficient mice had significantly elevated ATPase and ADPase activities in LN T cells. In a melanoma model, the growth of primary tumors and formation of metastasis were significantly attenuated in mice lacking CD73. Among tumor-infiltrating leukocytes there were fewer Tregs and mannose receptor-positive macrophages, and increased IFN-γ and NOS2 mRNA production in CD73-deficient mice. Treatment of tumor-bearing animals with soluble apyrase, an enzyme hydrolyzing ATP and ADP, significantly inhibited tumor growth and accumulation of intratumoral Tregs and mannose receptor-positive macrophages in the WT C57BL/6 mice but not in the CD73-deficient mice. Pharmacological inhibition of CD73 with α,ß-methylene-adenosine-5'-diphosphate in WT mice retarded tumor progression similarly to the genetic deletion of CD73. Together these data show that increased pericellular ATP degradation in the absence of CD73 activity in the host cells is a novel mechanism controlling anti-tumor immunity and tumor progression, and that the purinergic balance can be manipulated therapeutically to inhibit tumor growth.


Subject(s)
5'-Nucleotidase/physiology , Adenosine Triphosphatases/metabolism , Apyrase/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/genetics , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Apyrase/pharmacology , Cell Proliferation/drug effects , Disease Progression , Interferon-gamma/genetics , Lectins, C-Type/genetics , Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Macrophages/immunology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/genetics , Nitric Oxide Synthase Type II/genetics , Purines/metabolism , RNA, Messenger/genetics , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
8.
Endocr Relat Cancer ; 17(3): 785-96, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20576801

ABSTRACT

Reactive oxygen species, specifically hydrogen peroxide (H(2)O(2)), have a significant role in hormone production in thyroid tissue. Although recent studies have demonstrated that dual oxidases are responsible for the H(2)O(2) synthesis needed in thyroid hormone production, our data suggest a pivotal role for superoxide dismutase 3 (SOD3) as a major H(2)O(2)-producing enzyme. According to our results, Sod3 is highly expressed in normal thyroid, and becomes even more abundant in rat goiter models. We showed TSH-stimulated expression of Sod3 via phospholipase C-Ca(2+) and cAMP-protein kinase A, a pathway that might be disrupted in thyroid cancer. In line with this finding, we demonstrated an oncogene-dependent decrease in Sod3 mRNA expression synthesis in thyroid cancer cell models that corresponded to a similar decrease in clinical patient samples, suggesting that SOD3 could be used as a differentiation marker in thyroid cancer. Finally, the functional analysis in thyroid models indicated a moderate role for SOD3 in regulating normal thyroid cell proliferation being in line with our previous observations.


Subject(s)
Antigens, Differentiation/metabolism , Cell Differentiation , Superoxide Dismutase/metabolism , Thyroid Neoplasms/enzymology , Animals , Blotting, Western , Calcium/metabolism , Carcinoma , Carcinoma, Papillary , Cell Proliferation , Down-Regulation , Humans , Hydrogen Peroxide/metabolism , Male , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxides/metabolism , Thyroid Cancer, Papillary , Thyroid Carcinoma, Anaplastic , Thyroid Gland/enzymology , Thyroid Gland/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Tumor Cells, Cultured
9.
Cytotherapy ; 11(6): 726-37, 2009.
Article in English | MEDLINE | ID: mdl-19878059

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) have been used in a wide variety of pre-clinical experiments and in an increasing number of human clinical trials. Although many of these studies have shown different levels of engraftment, the exact fate of MSC after transplantation and the tissue response to their engraftment have not been investigated in detail. In the present work we studied the distribution of human MSC in a rat hind limb ischemic injury model immediately after transplantation and also analyzed the recipient tissue response to transplanted cells. METHODS: We tracked the in vivo fate of the transplanted MSC utilizing bioluminescence imaging, fluorescence microscopy and gene/protein expression analysis in a rat hind limb ischemia model. We also monitored the viability of transplanted cells by graft versus recipient expression analysis and determined the angiogenic and proliferative effect of transplantation by histologic staining. RESULTS: According to imaging analysis only a small portion of cells persisted for an extended period of time at the site of injury. Interestingly, recipient versus graft expression studies showed increased synthesis of rat-origin angiogenic factors and no human-origin mRNA or protein synthesis in transplanted tissues. More importantly, despite the lack of robust engraftment or growth factor secretion the transplantation procedure exerted a significant pro-angiogenic and pro-proliferative effect, which was mediated by angiogenic and mitogenic signaling pathways. CONCLUSIONS: Our results show an immediate temporal tissue effect in response to MSC transplantation that may represent a novel indirect paracrine mechanism for the beneficial effects of cell transplantation observed in injured tissues.


Subject(s)
Lower Extremity/blood supply , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic , Reperfusion Injury/surgery , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression/physiology , Graft vs Host Reaction/immunology , Host vs Graft Reaction/immunology , Humans , Lower Extremity/pathology , Lower Extremity/surgery , Male , Mesenchymal Stem Cells/cytology , Rats , Regeneration , Reperfusion Injury/pathology , Signal Transduction/physiology , Transduction, Genetic
10.
PLoS One ; 4(6): e5786, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19495415

ABSTRACT

Inflammatory cell migration characteristic of ischemic damages has a dual role providing the tissue with factors needed for tissue injury recovery simultaneously causing deleterious development depending on the quality and the quantity of infiltrated cells. Extracellular superoxide dismutase (SOD3) has been shown to have an anti-inflammatory role in ischemic injuries where it increases the recovery process by activating mitogen signal transduction and increasing cell proliferation. However, SOD3 derived effects on inflammatory cytokine and adhesion molecule expression, which would explain reduced inflammation in vascular lesions, has not been properly characterized. In the present work the effect of SOD3 on the inflammatory cell extravasation was studied in vivo in rat hind limb ischemia and mouse peritonitis models by identifying the migrated cells and analyzing SOD3-derived response on inflammatory cytokine and adhesion molecule expression. SOD3 overexpression significantly reduced TNFalpha, IL1alpha, IL6, MIP2, and MCP-1 cytokine and VCAM, ICAM, P-selectin, and E-selectin adhesion molecule expressions in injured tissues. Consequently the mononuclear cell, especially CD68+ monocyte and CD3+ T cell infiltration were significantly decreased whereas granulocyte migration was less affected. According to our data SOD3 has a selective anti-inflammatory role in ischemic damages preventing the migration of reactive oxygen producing monocyte/macrophages, which in excessive amounts could potentially further intensify the tissue injuries therefore suggesting potential for SOD3 in treatment of inflammatory disorders.


Subject(s)
Cytokines/biosynthesis , Inflammation , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/physiology , Animals , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement , Female , Granulocytes/cytology , Humans , Male , Mice , Mice, Inbred BALB C , Models, Biological , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism
11.
Mol Ther ; 17(3): 448-54, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19107121

ABSTRACT

Extracellular superoxide dismutase (SOD3) gene therapy has been shown to attenuate tissue damages and to improve the recovery of the tissue injuries, but the cellular events delivering the therapeutic response of the enzyme are not well defined. In the current work, we overexpressed SOD3 in rat hindlimb ischemia model to study the signal transduction and injury healing following the sod3 gene transfer. The data suggest a novel sod3 gene transfer-derived signal transduction cascade through Ras-Mek-Erk mitogenic pathway leading to activation of AP1 and CRE transcription factors, increased vascular endothelial growth factor (VEGF)-A and cyclin D1 expression, increased cell proliferation, and consequently improved metabolic functionality of the injured tissue. Increased cell proliferation could explain the improved metabolic performance and the healing of the tissue damages after the sod3 gene transfer. The present data is a novel description of the molecular mechanism of SOD3-mediated recovery of tissue injury and suggests a new physiological role for SOD3 as a Ras regulatory molecule in signal transduction.


Subject(s)
Extracellular Space/enzymology , Hindlimb/enzymology , Hindlimb/pathology , Ischemia/enzymology , Ischemia/pathology , Superoxide Dismutase/metabolism , Adenoviridae/genetics , Animals , Cell Line , Disease Models, Animal , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Hindlimb/injuries , Humans , Ischemia/genetics , MAP Kinase Signaling System , Male , Rabbits , Rats , Superoxide Dismutase/genetics , Transgenes/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL