Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 11(1): 722, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024825

ABSTRACT

Heterotopic ossification (HO) is an aberrant regenerative process with ectopic bone induction in response to musculoskeletal trauma, in which mesenchymal stem cells (MSC) differentiate into osteochondrogenic cells instead of myocytes or tenocytes. Despite frequent cases of hospitalized musculoskeletal trauma, the inflammatory responses and cell population dynamics that regulate subsequent wound healing and tissue regeneration are still unclear. Here we examine, using a mouse model of trauma-induced HO, the local microenvironment of the initial post-injury inflammatory response. Single cell transcriptome analyses identify distinct monocyte/macrophage populations at the injury site, with their dynamic changes over time elucidated using trajectory analyses. Mechanistically, transforming growth factor beta-1 (TGFß1)-producing monocytes/macrophages are associated with HO and aberrant chondrogenic progenitor cell differentiation, while CD47-activating peptides that reduce systemic macrophage TGFß levels and help ameliorate HO. Our data thus implicate CD47 activation as a therapeutic approach for modulating monocyte/macrophage phenotypes, MSC differentiation and HO formation during wound healing.


Subject(s)
Burns/pathology , Monocytes/pathology , Ossification, Heterotopic/pathology , Wound Healing/physiology , Animals , CD47 Antigen/metabolism , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Macrophages/pathology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice, Inbred C57BL , Mice, Transgenic , Peptides/pharmacology , Phagocytosis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
2.
Arterioscler Thromb Vasc Biol ; 39(11): 2353-2366, 2019 11.
Article in English | MEDLINE | ID: mdl-31644352

ABSTRACT

OBJECTIVE: Sepsis represents an acute life-threatening disorder resulting from a dysregulated host response. For patients who survive sepsis, there remains long-term consequences, including impaired inflammation, as a result of profound immunosuppression. The mechanisms involved in this long-lasting deficient immune response are poorly defined. Approach and Results: Sepsis was induced using the murine model of cecal ligation and puncture. Following a full recovery period from sepsis physiology, mice were subjected to our wound healing model and wound macrophages (CD11b+, CD3-, CD19-, Ly6G-) were sorted. Post-sepsis mice demonstrated impaired wound healing and decreased reepithelization in comparison to controls. Further, post-sepsis bone marrow-derived macrophages and wound macrophages exhibited decreased expression of inflammatory cytokines vital for wound repair (IL [interleukin]-1ß, IL-12, and IL-23). To evaluate if decreased inflammatory gene expression was secondary to epigenetic modification, we conducted chromatin immunoprecipitation on post-sepsis bone marrow-derived macrophages and wound macrophages. This demonstrated decreased expression of Mll1, an epigenetic enzyme, and impaired histone 3 lysine 4 trimethylation (activation mark) at NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells)-binding sites on inflammatory gene promoters in bone marrow-derived macrophages and wound macrophages from postcecal ligation and puncture mice. Bone marrow transplantation studies demonstrated epigenetic modifications initiate in bone marrow progenitor/stem cells following sepsis resulting in lasting impairment in peripheral macrophage function. Importantly, human peripheral blood leukocytes from post-septic patients demonstrate a significant reduction in MLL1 compared with nonseptic controls. CONCLUSIONS: These data demonstrate that severe sepsis induces stable mixed-lineage leukemia 1-mediated epigenetic modifications in the bone marrow, which are passed to peripheral macrophages resulting in impaired macrophage function and deficient wound healing persisting long after sepsis recovery.


Subject(s)
Epigenesis, Genetic , Inflammation/physiopathology , Macrophages/physiology , Sepsis/genetics , Sepsis/physiopathology , Wound Healing/physiology , Animals , Bone Marrow Cells/physiology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Immune Tolerance , Male , Mice, Inbred C57BL , Mice, Inbred Strains , Myeloid-Lymphoid Leukemia Protein/genetics , NF-kappa B/genetics , Promoter Regions, Genetic , Sepsis/metabolism
3.
J Immunol ; 199(5): 1865-1874, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28733487

ABSTRACT

Macrophages are critical immune cells for the clearance of microbial pathogens and cellular debris from peripheral tissues. Macrophage inflammatory responses are governed by gene expression patterns, and these patterns are often subject to epigenetic control. Chromatin modifications, such as histone methylation, regulate gene accessibility in macrophages, and macrophage polarization is governed in part by the expression and function of chromatin-modifying enzymes. The histone methyltransferase mixed-lineage leukemia 1 (MLL1) preferentially modifies lysine residue 4 on the unstructured protein tail of histone H3. MLL1 expression and function have been shown to be governed by signal transduction pathways that are activated by inflammatory stimuli, such as NF-κB. Therefore, we sought to investigate the role of MLL1 in mediating macrophage inflammatory responses. Bone marrow-derived macrophages from mice with a targeted MLL1 gene knockout (Lys2-Cre+/- MLL1fx/fx) exhibited decreased proinflammatory gene expression with concurrent decreases in activating histone methylation. However, MLL1-deficient macrophages also exhibited increased phagocytic and bacterial killing activity in vitro. RNA profiling of MLL1-knockout macrophages identified numerous genes involved with inflammatory responses whose expression was altered in response to TLR ligands or proinflammatory cytokines, including STAT4. STAT4-dependent cytokines, such as type I IFNs were able to drive MLL1 expression in macrophages, and MLL1-knockout macrophages exhibited decreased activating histone methylation in the STAT4 promoter. These results implicate an important role for MLL1-dependent epigenetic regulation of macrophage antimicrobial functions.


Subject(s)
Epigenesis, Genetic/immunology , Histone-Lysine N-Methyltransferase/metabolism , Infections/immunology , Macrophages/immunology , Myeloid-Lymphoid Leukemia Protein/metabolism , STAT4 Transcription Factor/metabolism , Animals , Bacteriolysis , Cells, Cultured , Chromatin Assembly and Disassembly , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , NF-kappa B/metabolism , STAT4 Transcription Factor/genetics , Signal Transduction , Transcriptome
4.
Cell Immunol ; 314: 63-72, 2017 04.
Article in English | MEDLINE | ID: mdl-28242024

ABSTRACT

Macrophage polarization plays a central role in both protective immunity and immunopathology. While the role of cytokines in driving macrophage polarization is well characterized, less is understood about the role of chemokines. The purpose of this study was to determine if CC chemokine 2 (CCL2/MCP1) could influence macrophage polarization in response to subsequent activation with cytokines and microbial products. Treatment of bone marrow-derived macrophages with CCL2 alone did not result in increased expression of either classical or alternatively-activated macrophage genes as compared to standard skewing cytokines or Toll-like receptor agonists. However, subsequent stimulation of CCL2 pre-treated macrophages with classical activation stimuli resulted in enhanced expression of genes associated with classical activation. This enhancement correlated with increased phosphorylation of ERK1/2 kinases, a decrease in expression of the ERK phosphatase Dusp6 and enhanced expression of miR-9. These results indicate that CCL2 supports the classical activation of macrophages, with miR-9 mediated down-regulation of Dusp6 and enhanced ERK-mediated signal transduction possibly mediating this enhanced pro-inflammatory gene expression.


Subject(s)
Dual Specificity Phosphatase 6/metabolism , Inflammation/immunology , Macrophage Activation , Macrophages/immunology , MicroRNAs/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chemokine CCL2/metabolism , Cytokines/metabolism , Dual Specificity Phosphatase 6/genetics , Female , Gene Expression Regulation , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...