Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Chest ; 152(2): 410-416, 2017 08.
Article in English | MEDLINE | ID: mdl-28130045

ABSTRACT

Airway eosinophilia is a hallmark of allergic asthma, and understanding mechanisms that promote increases in lung eosinophil numbers is important for effective pharmacotherapeutic development. It has become evident that expansion of hematopoietic compartments in the bone marrow (BM) promotes differentiation and trafficking of mature eosinophils to the airways. Hematopoietic progenitor cells egress the BM and home to the lungs, where in situ differentiation within the tissue provides an ongoing source of proinflammatory cells. In addition, hematopoietic progenitor cells in the airways can respond to locally derived alarmins to produce a panoply of cytokines, thereby themselves acting as effector proinflammatory cells that potentiate type 2 responses in eosinophilic asthma. In this review, we provide evidence for these findings and discuss novel targets for modulating eosinophilopoietic processes, migration, and effector function of precursor cells.


Subject(s)
Asthma/etiology , Hematopoiesis/physiology , Pulmonary Eosinophilia/etiology , Antibodies, Monoclonal, Humanized/therapeutic use , Asthma/drug therapy , Asthma/physiopathology , CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/physiology , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokines/antagonists & inhibitors , Cytokines/physiology , Eosinophils/cytology , Eosinophils/physiology , Hematopoietic Stem Cells/physiology , Humans , Membrane Proteins/antagonists & inhibitors , Pulmonary Eosinophilia/drug therapy , Pulmonary Eosinophilia/physiopathology , Respiratory Hypersensitivity/etiology , Respiratory Hypersensitivity/physiopathology , Thymic Stromal Lymphopoietin
2.
Shock ; 46(4): 390-7, 2016 10.
Article in English | MEDLINE | ID: mdl-27172159

ABSTRACT

BACKGROUND: Alternate erythropoietin (EPO)-mediated signaling via the EPOR/CD131 heteromeric receptor exerts the tissue-protective actions of EPO in a wide spectrum of injuries, especially ischemic diseases. Circulating endothelial progenitor cells contribute to endothelial repair and post-natal angiogenesis after chronic ischemic injury. This work aims to investigate the effects of ARA290, a specific agonist of EPOR/CD131 complex, on a subpopulation of endothelial progenitor cells named endothelial colony-forming cells (ECFCs) and to characterize its contribution to ECFCs-induced angiogenesis after peripheral ischemia. METHODS: ARA290 effects on ECFCs properties were studied using cell cultures in vitro. We injected ARA290 to mice undergoing chronic hindlimb ischemia (CLI) in combination with ECFC transplantation. The homing of transplanted ECFC to ischemic tissue in vivo was assessed by SPECT/CT imaging. RESULTS: In vitro, ARA290 enhanced the proliferation, migration, and resistance to H2O2-induced apoptosis of ECFCs. After ECFC transplantation to mice with CLI, a single ARA290 injection enhanced the ischemic/non-ischemic ratio of hindlimb blood flow and capillary density after 28 days and the homing of radiolabeled transplanted cells to the ischemic leg 4 h after transplantation. Prior neutralization of platelet-endothelial cell adhesion molecule-1 (CD31) expressed by the transplanted cells inhibited ARA290-induced improvement of homing. DISCUSSION: ARA290 induces specific improvement of the biological activity of ECFCs. ARA290 administration in combination with ECFCs has a synergistic effect on post-ischemic angiogenesis in vivo. This potentiation appears to rely, at least in part, on a CD31-dependent increase in homing of the transplanted cells to the ischemic tissue.


Subject(s)
Cytokine Receptor Common beta Subunit/metabolism , Oligopeptides/pharmacology , Receptors, Erythropoietin/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Hindlimb/metabolism , Hindlimb/pathology , Humans , Hydrogen Peroxide/pharmacology , Ischemia/metabolism , Ischemia/pathology , Mice , Oxidative Stress/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Receptors, Erythropoietin/agonists , Signal Transduction/drug effects , Single Photon Emission Computed Tomography Computed Tomography
3.
Immunity ; 36(6): 1031-46, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749353

ABSTRACT

GM-CSF (Csf-2) is a critical cytokine for the in vitro generation of dendritic cells (DCs) and is thought to control the development of inflammatory DCs and resident CD103(+) DCs in some tissues. Here we showed that in contrast to the current understanding, Csf-2 receptor acts in the steady state to promote the survival and homeostasis of nonlymphoid tissue-resident CD103(+) and CD11b(+) DCs. Absence of Csf-2 receptor on lung DCs abrogated the induction of CD8(+) T cell immunity after immunization with particulate antigens. In contrast, Csf-2 receptor was dispensable for the differentiation and innate function of inflammatory DCs during acute injuries. Instead, inflammatory DCs required Csf-1 receptor for their development. Thus, Csf-2 is important in vaccine-induced CD8(+) T cell immunity through the regulation of nonlymphoid tissue DC homeostasis rather than control of inflammatory DCs in vivo.


Subject(s)
Cytokine Receptor Common beta Subunit/physiology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Inflammation/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Lineage , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/deficiency , Cytokine Receptor Common beta Subunit/genetics , Dendritic Cells/classification , Dendritic Cells/cytology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endotoxemia/immunology , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Homeostasis , Lipopolysaccharides/toxicity , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/transplantation , Organ Specificity , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/immunology , Radiation Chimera , Spleen/immunology , Tamoxifen/pharmacology
4.
J Immunol ; 186(4): 1923-33, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21220694

ABSTRACT

GM-CSF is an important cytokine involved in myeloid differentiation and inflammatory processes. Signaling through the GM-CSFR also plays a critical role in the generation of monocyte-derived dendritic cells (DC). In this article, we report that the Src-like adaptor protein (SLAP) functions as a negative regulator of the GM-CSFR. In bone marrow-derived DC (BM-DC) lacking SLAP and the closely related SLAP2, downregulation of GM-CSFRß is impaired, leading to enhanced phosphorylation of Jak2 and prolonged activation of Akt and Erk1/2 in response to GM-CSF stimulation. Compared with wild-type bone marrow, SLAP/SLAP2(-/-) bone marrow gave rise to similar numbers of CD11c(+) and CD11b(+) DC, but SLAP/SLAP2(-/-) BM-DC failed to acquire high levels of MHC class II, CD80, and CD86, indicating an impairment in maturation. Furthermore, MHC class II expression in SLAP/SLAP2(-/-) BM-DC was rescued by decreasing GM-CSF concentration, suggesting that enhanced GM-CSF signaling mediates the block in maturation. In addition, SLAP/SLAP2(-/-) BM-DC produced less IL-12 and TNF-α in response to LPS compared with controls and failed to stimulate T cells in an MLR. Ag-specific T cell activation assays showed that SLAP/SLAP2(-/-) BM-DC were less robust at inducing IFN-γ secretion by DO11.10 T cells. These results indicated that SLAP-mediated GM-CSFR regulation is important for the generation of functionally mature monocytic DC.


Subject(s)
Cell Differentiation/immunology , Cytokine Receptor Common beta Subunit/physiology , Dendritic Cells/immunology , Growth Inhibitors/physiology , Monocytes/immunology , Proto-Oncogene Proteins pp60(c-src)/physiology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Cell Differentiation/genetics , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Down-Regulation/genetics , Down-Regulation/immunology , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Mice , Mice, Knockout , Mice, Transgenic , Monocytes/metabolism , Monocytes/pathology , Proto-Oncogene Proteins pp60(c-src)/deficiency , Proto-Oncogene Proteins pp60(c-src)/genetics , Signal Transduction/genetics
5.
J Immunol ; 185(3): 1606-15, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20622119

ABSTRACT

GM-CSF, IL-3, and IL-5 are proinflammatory cytokines that control the production and function of myeloid and lymphoid cells. Their receptors are composed of a ligand-specific alpha subunit and a shared common signal-transducing beta subunit (beta common receptor or GM-CSFR beta [beta(c)]). The pleiotropic nature of biologic outcomes mediated by beta(c) and the presence of large, uncharacterized regions of its cytoplasmic domain suggest that much remains to be learned about its downstream signaling pathways. Although some previous work has attempted to link beta(c) with NF-kappaB activation, a definitive mechanism that mediates this pathway has not been described and, to date, it has not been clear whether the receptor can directly activate NF-kappaB. We demonstrate that NF-kappaB activation by beta(c) is dependent on TNFR-associated factor 6 (TRAF6) and that association of TRAF6 with beta(c) requires a consensus-binding motif found in other molecules known to interact with TRAF6. Furthermore, point mutation of this motif abrogated the ability of beta(c) to mediate NF-kappaB activation and reduced the viability of an IL-3-dependent hematopoietic cell line. Because this receptor plays a key role in hematopoiesis and the beta(c) cytoplasmic domain identified in this work mediates hematopoietic cell viability, this new pathway is likely to contribute to immune cell biology. This work is significant because it is the first description of a TRAF6-dependent signaling pathway associated with a type I cytokine receptor. It also suggests that TRAF6, a mediator of TNFR and TLR signaling, may be a common signaling intermediate in diverse cytokine receptor systems.


Subject(s)
Cytokine Receptor Common beta Subunit/physiology , NF-kappa B/physiology , TNF Receptor-Associated Factor 6/metabolism , Active Transport, Cell Nucleus/immunology , Animals , Cells, Cultured , Clone Cells , Consensus Sequence , Cytokine Receptor Common beta Subunit/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Humans , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Biosynthesis/immunology , Protein Structure, Tertiary/genetics , Protein Transport/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , TNF Receptor-Associated Factor 6/deficiency , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...