7 research outputs found

    Circulating Fibrocytes Prepare the Lung for Cancer Metastasis by Recruiting Ly-6C+ Monocytes Via CCL2

    Get PDF
    Fibrocytes are circulating, hematopoietic cells that express CD45 and Col1a1. They contribute to wound healing and several fibrosing disorders by mechanisms that are poorly understood. In this report, we demonstrate that fibrocytes predispose the lung to B16-F10 metastasis by recruiting Ly-6C+ monocytes. To do so, we isolated fibrocytes expressing CD45, CD11b, CD13, and Col1a1 from the lungs of wild type (WT) and Ccr5−/− mice. WT but not Ccr5−/− fibrocytes increased the number of metastatic foci when injected into Ccr5−/− mice (73 ± 2 versus 32 ± 5; p < 0.001). This process was MMP9 dependent. Injection of WT enhanced GFP+ fibrocytes also increased the number of Gr-1Int, CD11b+, and enhanced GFP− monocytes. Like premetastatic-niche monocytes, these recruited cells expressed Ly-6C, CD117, and CD45. The transfer of these cells into Ccr5−/− mice enhanced metastasis (90 ± 8 foci) compared with B cells (27 ± 2), immature dendritic cells (31 ± 6), or alveolar macrophages (28 ± 3; p < 0.05). WT and Ccl2−/− fibrocytes also stimulated Ccl2 expression in the lung by 2.07 ± 0.05- and 2.78 ± 0.36-fold compared with Ccr5−/− fibrocytes (1.0 ± 0.06; p < 0.05). Furthermore, WT fibrocytes did not increase Ly-6C+ monocytes in Ccr2−/− mice and did not promote metastasis in either Ccr2−/− or Ccl2−/− mice. These data support our hypothesis that fibrocytes contribute to premetastatic conditioning by recruiting Ly-6C+ monocytes in a chemokine-dependent process. This work links metastatic risk to conditions that mobilize fibrocytes, such as inflammation and wound repair

    C-C Chemokine Receptor 5 on Pulmonary Mesenchymal Cells Promotes Experimental Metastasis via the Induction of Erythroid Differentiation Regulator 1

    Get PDF
    C-C Chemokine receptor five knockout (Ccr5-/-) mice develop fewer experimental pulmonary metastases than wild type (WT) mice. This phenomenon was explored by applying gene-expression profiling to the lungs of mice with these metastases. Consequently, Erythroid Differentiation Regulator 1 (Erdr1) was identified as upregulated in the WT mice. Though commonly associated with bone marrow stroma, Erdr1 was differentially expressed in WT pulmonary mesenchymal cells (PMCs) and murine embryonic fibroblasts (MEFs). Moreover, the Ccr5 ligand Ccl4 increased its expression by 3.36 ± 0.14 fold. Ccr5 signaling was dependent on the Map2k but not the Pi3k pathway since treatment with U0126 inhibited upregulation of Erdr1 but treatment with LY294002 increased the expression by 3.44 ± 0.92 fold (p < 0.05). Erdr1's effect on B16-F10 melanoma metastasis was verified by the adoptive transfer of WT MEFs into Ccr5-/- mice. In this model, MEFs that had been transduced with Erdr1 shRNA lowered metastasis by 33% compared to control transduced MEFs. The relevance of ERDR1 on human disease was assessed by co-culturing chronic lymphocytic leukemia (CLL) cells with M2-10B4 stromal cells that had been transfected with shRNA or control plasmids. After 96 hours of co-culture, the cell counts were higher with control cell lines compared with Erdr1 knockdown lines (OR 1.88 ± 0.27, 2.52 ± 0.66 respectively). This increase was associated with a decrease in apoptotic cells (OR 0.69 ± 0.18, 0.58 ± 0.12 respectively)

    Circulating Fibrocytes Prepare the Lung for Cancer Metastasis by Recruiting Ly-6C+ Monocytes Via CCL2

    Get PDF
    Fibrocytes are circulating, hematopoietic cells that express CD45 and Col1a1. They contribute to wound healing and several fibrosing disorders by mechanisms that are poorly understood. In this report, we demonstrate that fibrocytes predispose the lung to B16-F10 metastasis by recruiting Ly-6C+ monocytes. To do so, we isolated fibrocytes expressing CD45, CD11b, CD13, and Col1a1 from the lungs of wild type (WT) and Ccr52/2 mice. WT but not Ccr52/2 fibrocytes increased the number of metastatic foci when injected into Ccr52/2 mice (73 6 2 versus 32 6 5; p < 0.001). This process was MMP9 dependent. Injection of WT enhanced GFP+ fibrocytes also increased the number of Gr-1Int, CD11b+, and enhanced GFP\u2013 monocytes. Like premetastaticniche monocytes, these recruited cells expressed Ly-6C, CD117, and CD45. The transfer of these cells into Ccr52/2 mice enhanced metastasis (90 6 8 foci) compared with B cells (27 6 2), immature dendritic cells (31 6 6), or alveolar macrophages (28 6 3; p <0.05). WTand Ccl22/2 fibrocytes also stimulated Ccl2 expression in the lung by 2.07 6 0.05- and 2.78 6 0.36-fold compared with Ccr52/2 fibrocytes (1.0 6 0.06; p < 0.05). Furthermore, WT fibrocytes did not increase Ly-6C+ monocytes in Ccr22/2 mice and did not promote metastasis in either Ccr22/2 or Ccl22/2 mice. These data support our hypothesis that fibrocytes contribute to premetastatic conditioning by recruiting Ly-6C+ monocytes in a chemokine-dependent process. This work links metastatic risk to conditions that mobilize fibrocytes, such as inflammation and wound repair

    C-C Chemokine Receptor 5 on Pulmonary Mesenchymal Cells Promotes Experimental Metastasis via the Induction of Erythroid Differentiation Regulator 1

    No full text
    C-C Chemokine receptor five knockout (Ccr5(-/-)) mice develop fewer experimental pulmonary metastases than wild type (WT) mice. This phenomenon was explored by applying gene-expression profiling to the lungs of mice with these metastases. Consequently, Erythroid Differentiation Regulator 1 (Erdr1) was identified as upregulated in the WT mice. Though commonly associated with bone marrow stroma, Erdr1 was differentially expressed in WT pulmonary mesenchymal cells (PMCs) and murine embryonic fibroblasts (MEFs). Moreover, the Ccr5 ligand Ccl4 increased its expression by 3.36 ± 0.14 fold. Ccr5 signaling was dependent on the Map2k but not the Pi3k pathway since treatment with U0126 inhibited upregulation of Erdr1 but treatment with LY294002 increased the expression by 3.44 ± 0.92 fold (p < 0.05). Erdr1's effect on B16-F10 melanoma metastasis was verified by the adoptive transfer of WT MEFs into Ccr5(-/-) mice. In this model, MEFs that had been transduced with Erdr1 shRNA lowered metastasis by 33% compared to control transduced MEFs. The relevance of ERDR1 on human disease was assessed by co-culturing chronic lymphocytic leukemia (CLL) cells with M2-10B4 stromal cells that had been transfected with shRNA or control plasmids. After 96 hours of co-culture, the cell counts were higher with control cell lines compared with Erdr1 knockdown lines (OR 1.88 ± 0.27, 2.52 ± 0.66 respectively). This increase was associated with a decrease in apoptotic cells (OR 0.69 ± 0.18, 0.58 ± 0.12 respectively). IMPLICATIONS: Therefore, ERDR1 is a stromal-derived factor that promotes cancer cell survival in vitro and in an experimental metastasis model

    Deletion of the Protein Kinase A/Protein Kinase G Target SMTNL1 Promotes an Exercise-adapted Phenotype in Vascular Smooth Muscle*S⃞

    Get PDF
    In vivo protein kinases A and G (PKA and PKG) coordinately phosphorylate a broad range of substrates to mediate their various physiological effects. The functions of many of these substrates have yet to be defined genetically. Herein we show a role for smoothelin-like protein 1 (SMTNL1), a novel in vivo target of PKG/PKA, in mediating vascular adaptations to exercise. Aortas from smtnl1-/- mice exhibited strikingly enhanced vasorelaxation before exercise, similar in extent to that achieved after endurance training of wild-type littermates. Additionally, contractile responses to α-adrenergic agonists were greatly attenuated. Immunological studies showed SMTNL1 is expressed in smooth muscle and type 2a striated muscle fibers. Consistent with a role in adaptations to exercise, smtnl1-/- mice also exhibited increased type 2a fibers before training and better performance after forced endurance training compared smtnl1+/+ mice. Furthermore, exercise was found to reduce expression of SMTNL1, particularly in female mice. In both muscle types, SMTNL1 is phosphorylated at Ser-301 in response to adrenergic signals. In vitro SMTNL1 suppresses myosin phosphatase activity through a substrate-directed effect, which is relieved by Ser-301 phosphorylation. Our findings suggest roles for SMTNL1 in cGMP/cAMP-mediated adaptations to exercise through mechanisms involving direct modulation of contractile activity

    Deletion of the Protein Kinase A/Protein Kinase G Target SMTNL1 Promotes an Exercise-adapted Phenotype in Vascular Smooth Muscle

    No full text
    In vivo protein kinases A and G (PKA and PKG) coordinately phosphorylate a broad range of substrates to mediate their various physiological effects. The functions of many of these substrates have yet to be defined genetically. Herein we show a role for smoothelin-like protein 1 (SMTNL1) a novel in vivo target of PKG/PKA in mediating vascular adaptations to exercise. Aortas from smtnl1-/- mice exhibited strikingly enhanced vasorelaxation before exercise similar in extent to that achieved after endurance training of wild-type littermates. Additionally con- tractile responses to alpha-adrenergic agonists were greatly attenuated. Immunological studies showed SMTNL1 is expressed in smooth muscle and type 2a striated muscle fibers. Consistent with a role in adaptations to exercise smtnl1-/- mice also exhibited increased type 2a fibers before training and better performance after forced endurance training compared smtnl1+/+ mice. Furthermore exercise was found to reduce expression of SMTNL1 particularly in female mice. In both muscle types SMTNL1 is phosphorylated at Ser-301 in response to adrenergic signals. In vitro SMTNL1 suppresses myosin phosphatase activity through a substrate-directed effect which is relieved by Ser- 301 phosphorylation. Our findings suggest roles for SMTNL1 in cGMP/cAMP-mediated adaptations to exercise through mechanisms involving direct modulation of contractile activity. Originally published Journal of Biological Chemistry Vol. 283 No. 17 Apr 200
    corecore