Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Rep ; 41(11): 111797, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36516754

ABSTRACT

Persistent neutrophil-dominated lung inflammation contributes to lung damage in cystic fibrosis (CF). However, the mechanisms that drive persistent lung neutrophilia and tissue deterioration in CF are not well characterized. Starting from the observation that, in patients with CF, c-c motif chemokine receptor 2 (CCR2)+ monocytes/macrophages are abundant in the lungs, we investigate the interplay between monocytes/macrophages and neutrophils in perpetuating lung tissue damage in CF. Here we show that CCR2+ monocytes in murine CF lungs drive pathogenic transforming growth factor ß (TGF-ß) signaling and sustain a pro-inflammatory environment by facilitating neutrophil recruitment. Targeting CCR2 to lower the numbers of monocytes in CF lungs ameliorates neutrophil inflammation and pathogenic TGF-ß signaling and prevents lung tissue damage. This study identifies CCR2+ monocytes as a neglected contributor to the pathogenesis of CF lung disease and as a therapeutic target for patients with CF, for whom lung hyperinflammation and tissue damage remain an issue despite recent advances in CF transmembrane conductance regulator (CFTR)-specific therapeutic agents.


Subject(s)
Cystic Fibrosis , Pneumonia , Humans , Mice , Animals , Cystic Fibrosis/pathology , Monocytes/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator , Pneumonia/pathology , Lung/pathology , Inflammation/pathology , Receptors, Chemokine/metabolism , Macrophages/metabolism , Transforming Growth Factor beta/metabolism
2.
Exp Mol Med ; 54(5): 639-652, 2022 05.
Article in English | MEDLINE | ID: mdl-35581352

ABSTRACT

Overwhelming neutrophilic inflammation is a leading cause of lung damage in many pulmonary diseases, including cystic fibrosis (CF). The heme oxygenase-1 (HO-1)/carbon monoxide (CO) pathway mediates the resolution of inflammation and is defective in CF-affected macrophages (MΦs). Here, we provide evidence that systemic administration of PP-007, a CO releasing/O2 transfer agent, induces the expression of HO-1 in a myeloid differentiation factor 88 (MyD88) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)-dependent manner. It also rescues the reduced HO-1 levels in CF-affected cells induced in response to lipopolysaccharides (LPS) or Pseudomonas aeruginosa (PA). Treatment of CF and muco-obstructive lung disease mouse models with a single clinically relevant dose of PP-007 leads to effective resolution of lung neutrophilia and to decreased levels of proinflammatory cytokines in response to LPS. Using HO-1 conditional knockout mice, we show that the beneficial effect of PP-007 is due to the priming of circulating monocytes trafficking to the lungs in response to infection to express high levels of HO-1. Finally, we show that PP-007 does not compromise the clearance of PA in the setting of chronic airway infection. Overall, we reveal the mechanism of action of PP-007 responsible for the immunomodulatory function observed in clinical trials for a wide range of diseases and demonstrate the potential use of PP-007 in controlling neutrophilic pulmonary inflammation by promoting the expression of HO-1 in monocytes/macrophages.


Subject(s)
Cystic Fibrosis , Pneumonia , Animals , Cystic Fibrosis/complications , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Heme Oxygenase-1 , Inflammation/metabolism , Lipopolysaccharides/metabolism , Lung/pathology , Mice , Monocytes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pneumonia/pathology
3.
RSC Adv ; 11(15): 8664-8673, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-35423352

ABSTRACT

Organic polymer/inorganic particle composites with thermoelectric (TE) properties have witnessed rapid progress in recent years. Nevertheless, both development of novel polymers and optimization of compositing methods remain highly desirable. In this study, we first demonstrated a simulated in situ coagulation strategy for construction of high-performance thermoelectric materials by utilizing single-walled carbon nanotubes (SWCNTs) and a new D-A polymer TPO-TTP12 that was synthesized via incorporating dioxothiopyrone subunit into a polymeric chain. It was proven that the preparation methods have a significant influence on thermoelectric properties of the TPO-TTP12/SWCNT composites. The in situ prepared composite films tend to achieve much better thermoelectric performances than those prepared by simply mixing the corresponding polymer with SWCNTs. As a result, the in situ compositing obtains the highest Seebeck coefficient of 66.10 ± 0.05 µV K-1 at the TPO-TTP12-to-SWCNT mass ratio of 1/2, and the best electrical conductivity of up to 500.5 ± 53.3 S cm-1 at the polymer/SWCNT mass ratio of 1/20, respectively; moreover, the power factor for the in situ prepared composites reaches a maximum value of 141.94 ± 1.47 µW m-1 K-2, far higher than that of 104.68 ± 0.86 µW m-1 K-2 for the by-mixing produced composites. This indicates that the dioxothiopyrone moiety is a promising building block for constructing thermoelectric polymers, and the simulated in situ compositing strategy is a promising way to improve TE properties of composite materials.

4.
Blood ; 134(18): 1547-1557, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31439541

ABSTRACT

The mechanisms underlying thrombocytosis in patients with iron deficiency anemia remain unknown. Here, we present findings that support the hypothesis that low iron biases the commitment of megakaryocytic (Mk)-erythroid progenitors (MEPs) toward the Mk lineage in both human and mouse. In MEPs of transmembrane serine protease 6 knockout (Tmprss6-/-) mice, which exhibit iron deficiency anemia and thrombocytosis, we observed a Mk bias, decreased labile iron, and decreased proliferation relative to wild-type (WT) MEPs. Bone marrow transplantation assays suggest that systemic iron deficiency, rather than a local role for Tmprss6-/- in hematopoietic cells, contributes to the MEP lineage commitment bias observed in Tmprss6-/- mice. Nontransgenic mice with acquired iron deficiency anemia also show thrombocytosis and Mk-biased MEPs. Gene expression analysis reveals that messenger RNAs encoding genes involved in metabolic, vascular endothelial growth factor, and extracellular signal-regulated kinase (ERK) pathways are enriched in Tmprss6-/- vs WT MEPs. Corroborating our findings from the murine models of iron deficiency anemia, primary human MEPs exhibit decreased proliferation and Mk-biased commitment after knockdown of transferrin receptor 2, a putative iron sensor. Signal transduction analyses reveal that both human and murine MEP have lower levels of phospho-ERK1/2 in iron-deficient conditions compared with controls. These data are consistent with a model in which low iron in the marrow environment affects MEP metabolism, attenuates ERK signaling, slows proliferation, and biases MEPs toward Mk lineage commitment.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Cell Differentiation/physiology , Megakaryocyte Progenitor Cells/metabolism , Megakaryocytes/metabolism , Anemia, Iron-Deficiency/complications , Animals , Cell Proliferation , Humans , Iron , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Mice , Mice, Knockout , Thrombocytosis/etiology , Thrombocytosis/metabolism
5.
Thromb Haemost ; 119(5): 744-757, 2019 May.
Article in English | MEDLINE | ID: mdl-30861547

ABSTRACT

Cadherins play a major role in mediating cell-cell adhesion, which shares many parallels with platelet-platelet interactions during aggregate formation and clot stabilization. Platelets express epithelial (E)-cadherin, but its contribution to platelet function and/or platelet production is currently unknown. To assess the role of E-cadherin in platelet production and function in vitro and in vivo, we utilized a megakaryocyte-specific E-cadherin knockout mouse model. Loss of E-cadherin in megakaryocytes does not affect megakaryocyte maturation, platelet number or size. However, platelet dysfunction in the absence of E-cadherin is revealed when conditional knockout mice are challenged with acute antibody-mediated platelet depletion. Unlike wild-type mice that recover fully, knockout mice die within 72 hours post-antibody administration, likely from haemorrhage. Furthermore, conditional knockout mice have prolonged tail bleeding times, unstable clot formation, reduced clot retraction and reduced fibrin deposition in in vivo injury models. Murine platelet aggregation in vitro in response to thrombin and thrombin receptor activating peptide is compromised in E-cadherin null platelets, while aggregation in response to adenosine diphosphate (ADP) is not significantly different. Consistent with this, in vitro aggregation of primary human platelets in response to thrombin is decreased by an inhibitory E-cadherin antibody. Integrin activation and granule secretion in response to ADP and thrombin are not affected in E-cadherin null platelets, but Akt and glycogen synthase kinase 3ß (GSK3ß) activation are attenuated, suggesting a that E-cadherin contributes to aggregation, clot stabilization and retraction that is mediated by phosphoinositide 3-kinase/Akt/GSK3ß signalling. In summary, E-cadherin plays a salient role in platelet aggregation and clot stability.


Subject(s)
Blood Platelets/physiology , Cadherins/metabolism , Liver/pathology , Megakaryocytes/physiology , Thrombosis/metabolism , Animals , Bleeding Time , Blood Coagulation , Cadherins/genetics , Cell Adhesion , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Platelet Aggregation , Signal Transduction , Thrombin/metabolism
7.
Cell Rep ; 25(8): 2083-2093.e4, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30463007

ABSTRACT

Megakaryocytic-erythroid progenitors (MEPs) give rise to the cells that produce red blood cells and platelets. Although the mechanisms underlying megakaryocytic (MK) and erythroid (E) maturation have been described, those controlling their specification from MEPs are unknown. Single-cell RNA sequencing of primary human MEPs, common myeloid progenitors (CMPs), megakaryocyte progenitors, and E progenitors revealed a distinct transitional MEP signature. Inferred regulatory transcription factors (TFs) were associated with differential expression of cell cycle regulators. Genetic manipulation of selected TFs validated their role in lineage specification and demonstrated coincident modulation of the cell cycle. Genetic and pharmacologic modulation demonstrated that cell cycle activation is sufficient to promote E versus MK specification. These findings, obtained from healthy human cells, lay a foundation to study the mechanisms underlying benign and malignant disease states of the megakaryocytic and E lineages.


Subject(s)
Cell Cycle , Cell Lineage , Megakaryocyte-Erythroid Progenitor Cells/cytology , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Gene Regulatory Networks , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Proto-Oncogene Proteins c-myc/metabolism , Reproducibility of Results , Signal Transduction , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 314(5): L882-L892, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29345196

ABSTRACT

Surfactant protein C (SPC), a key component of pulmonary surfactant, also plays a role in regulating inflammation. SPC deficiency in patients and mouse models is associated with increased inflammation and delayed repair, but the key drivers of SPC-regulated inflammation in response to injury are largely unknown. This study focuses on a new mechanism of SPC as an anti-inflammatory molecule using SPC-TK/SPC-KO (surfactant protein C-thymidine kinase/surfactant protein C knockout) mice, which represent a novel sterile injury model that mimics clinical acute respiratory distress syndrome (ARDS). SPC-TK mice express the inducible suicide gene thymidine kinase from by the SPC promoter, which targets alveolar type 2 (AT2) cells for depletion in response to ganciclovir (GCV). We compared GCV-induced injury and repair in SPC-TK mice that have normal endogenous SPC expression with SPC-TK/SPC-KO mice lacking SPC expression. In contrast to SPC-TK mice, SPC-TK/SPC-KO mice treated with GCV exhibited more severe inflammation, resulting in over 90% mortality; there was only 8% mortality of SPC-TK animals. SPC-TK/SPC-KO mice had highly elevated inflammatory cytokines and granulocyte infiltration in the bronchoalveolar lavage (BAL) fluid. Consistent with a proinflammatory phenotype, immunofluorescence revealed increased phosphorylated signal transduction and activation of transcription 3 (pSTAT3), suggesting enhanced Janus kinase (JAK)/STAT activation in inflammatory and AT2 cells of SPC-TK/SPC-KO mice. The level of suppressor of cytokine signaling 3, an anti-inflammatory mediator that decreases pSTAT3 signaling, was significantly decreased in the BAL fluid of SPC-TK/SPC-KO mice. Hyperactivation of pSTAT3 and inflammation were rescued by AZD1480, a JAK1/2 inhibitor. Our findings showing a novel role for SPC in regulating inflammation via JAK/STAT may have clinical applications.


Subject(s)
Disease Models, Animal , Janus Kinase 1/metabolism , Lung Injury/prevention & control , Peptides/physiology , Pneumonia/prevention & control , STAT3 Transcription Factor/metabolism , Thymidine Kinase/physiology , Animals , Intercellular Signaling Peptides and Proteins , Janus Kinase 1/genetics , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Surfactant-Associated Protein C , STAT3 Transcription Factor/genetics
9.
Sci Rep ; 7(1): 10882, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28883468

ABSTRACT

Macrophages (MΦs) with mutations in cystic fibrosis transmembrane conductance regulator (CFTR) have blunted induction of PI3K/AKT signaling in response to TLR4 activation, leading to hyperinflammation, a hallmark of cystic fibrosis (CF) disease. Here, we show that Ezrin links CFTR and TLR4 signaling, and is necessary for PI3K/AKT signaling induction in response to MΦ activation. Because PI3K/AKT signaling is critical for immune regulation, Ezrin-deficient MΦs are hyperinflammatory and have impaired Pseudomonas aeruginosa phagocytosis, phenocopying CF MΦs. Importantly, we show that activated CF MΦs have reduced protein levels and altered localization of the remaining Ezrin to filopodia that form during activation. In summary, we have described a direct link from CFTR to Ezrin to PI3K/AKT signaling that is disrupted in CF, and thus promotes hyper-inflammation and weakens phagocytosis.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/pathology , Cytoskeletal Proteins/metabolism , Macrophage Activation , Macrophages/immunology , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Cystic Fibrosis/complications , Disease Models, Animal , Mice , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/immunology
10.
PLoS One ; 12(5): e0178095, 2017.
Article in English | MEDLINE | ID: mdl-28542600

ABSTRACT

Genome-wide association studies have identified a genetic variant at 3p14.3 (SNP rs1354034) that strongly associates with platelet number and mean platelet volume in humans. While originally proposed to be intronic, analysis of mRNA expression in primary human hematopoietic subpopulations reveals that this SNP is located directly upstream of the predominantly expressed ARHGEF3 isoform in megakaryocytes (MK). We found that ARHGEF3, which encodes a Rho guanine exchange factor, is dramatically upregulated during both human and murine MK maturation. We show that the SNP (rs1354034) is located in a DNase I hypersensitive region in human MKs and is an expression quantitative locus (eQTL) associated with ARHGEF3 expression level in human platelets, suggesting that it may be the causal SNP that accounts for the variations observed in human platelet traits and ARHGEF3 expression. In vitro human platelet activation assays revealed that rs1354034 is highly correlated with human platelet activation by ADP. In order to test whether ARHGEF3 plays a role in MK development and/or platelet function, we developed an Arhgef3 KO/LacZ reporter mouse model. Reflecting changes in gene expression, LacZ expression increases during MK maturation in these mice. Although Arhgef3 KO mice have significantly larger platelets, loss of Arhgef3 does not affect baseline MK or platelets nor does it affect platelet function or platelet recovery in response to antibody-mediated platelet depletion compared to littermate controls. In summary, our data suggest that modulation of ARHGEF3 gene expression in humans with a promoter-localized SNP plays a role in human MKs and human platelet function-a finding resulting from the biological follow-up of human genetic studies. Arhgef3 KO mice partially recapitulate the human phenotype.


Subject(s)
Blood Platelets/metabolism , Polymorphism, Single Nucleotide , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Blood Platelets/cytology , Cell Differentiation/physiology , Cell Size , Cohort Studies , Female , Fetal Blood , Gene Expression Regulation , Humans , Male , Mean Platelet Volume , Megakaryocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic
11.
Blood ; 128(7): 923-33, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27268089

ABSTRACT

Bipotent megakaryocyte/erythroid progenitors (MEPs) give rise to progeny limited to the megakaryocyte (Mk) and erythroid (E) lineages. We developed a novel dual-detection functional in vitro colony-forming unit (CFU) assay for single cells that differentiates down both the Mk and E lineages (CFU-Mk/E), which allowed development and validation of a novel purification strategy for the identification and quantitation of primary functional human MEPs from granulocyte colony-stimulating factor-mobilized peripheral blood and bone marrow. Applying this assay to fluorescence-activated cell sorter-sorted cell populations, we found that the Lin(-)CD34(+)CD38(mid)CD45RA(-)FLT3(-)MPL(+)CD36(-)CD41(-) population is much more highly enriched for bipotent MEPs than any previously reported subpopulations. We also developed purification strategies for primary human lineage-committed Mk and E progenitors identified as CFU-Mk and burst forming unit-E. Comparative expression analyses in MEP, MkP, and ErP populations revealed differential expression of MYB We tested whether alterations in MYB concentration affect the Mk-E fate decision at the single cell level in MEPs and found that short hairpin RNA-mediated MYB knockdown promoted commitment of MEPs to the Mk lineage, further defining its role in MEP lineage fate. There are numerous applications for these novel enrichment strategies, including facilitating mechanistic studies of MEP lineage commitment, improving approaches for in vitro expansion of Mk and E cells, and developing improved therapies for benign and malignant hematologic disease.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Megakaryocyte-Erythroid Progenitor Cells/cytology , Adult , Cell Lineage , Cell Separation , Colony-Forming Units Assay , Erythroid Cells/cytology , Erythroid Cells/metabolism , Humans , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocytes/cytology , Phenotype , Proto-Oncogene Proteins c-myb/metabolism , Receptors, Thrombopoietin/metabolism , fms-Like Tyrosine Kinase 3/metabolism
12.
Thromb Haemost ; 116(3): 506-16, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27345948

ABSTRACT

Leukemia-Associated RhoGEF (LARG) is highly expressed in platelets, which are essential for maintaining normal haemostasis. We studied the function of LARG in murine and human megakaryocytes and platelets with Larg knockout (KO), shRNA-mediated knockdown and small molecule-mediated inhibition. We found that LARG is important for human, but not murine, megakaryocyte maturation. Larg KO mice exhibit macrothrombocytopenia, internal bleeding in the ovaries and prolonged bleeding times. KO platelets have impaired aggregation, α-granule release and integrin α2bß3 activation in response to thrombin and thromboxane, but not to ADP. The same agonist-specific reductions in platelet aggregation occur in human platelets treated with a LARG inhibitor. Larg KO platelets have reduced RhoA activation and myosin light chain phosphorylation, suggesting that Larg plays an agonist-specific role in platelet signal transduction. Using two different in vivo assays, Larg KO mice are protected from in vivo thrombus formation. Together, these results establish that LARG regulates human megakaryocyte maturation, and is critical for platelet function in both humans and mice.


Subject(s)
Blood Platelets/metabolism , Rho Guanine Nucleotide Exchange Factors/blood , rho GTP-Binding Proteins/blood , rhoA GTP-Binding Protein/blood , Animals , Bleeding Time , Blood Platelets/drug effects , Gene Knockdown Techniques , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Light Chains/blood , Platelet Function Tests , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/deficiency , Rho Guanine Nucleotide Exchange Factors/genetics , Thrombin/metabolism , Thrombin/pharmacology , Thrombopoiesis/genetics , Thrombopoiesis/physiology , Thromboxanes/blood , Thromboxanes/pharmacology , rho GTP-Binding Proteins/agonists , rhoA GTP-Binding Protein/agonists
13.
Am J Physiol Lung Cell Mol Physiol ; 310(8): L711-9, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26851259

ABSTRACT

Cystic fibrosis (CF) is caused by homozygous mutations of the CF transmembrane conductance regulator (CFTR) Cl(-) channel, which result in chronic pulmonary infection and inflammation, the major cause of morbidity and mortality. Although these processes are clearly related to each other, each is likely to contribute to the pathology differently. Understanding the contribution of each of these processes to the overall pathology has been difficult, because they are usually so intimately connected. Various CF mouse models have demonstrated abnormal immune responses compared with wild-type (WT) littermates when challenged with live bacteria or bacterial products acutely. However, these studies have not investigated the consequences of persistent inflammation on lung tissue in CF mice, which may better model the lung pathology in patients. We characterized the lung pathology and immune response of Cftr(-/-) (CF) and Cftr(+/+) (WT) mice to chronic administration of Pseudomonas aeruginosa lipopolysaccharide (LPS). We show that, after long-term repeated LPS exposure, CF mice develop an abnormal and persistent immune response, which is associated with more robust structural changes in the lung than those observed in WT mice. Although CF mice and their WT littermates develop lung pathology after chronic exposure to LPS, the inflammation and damage resolve in WT mice. However, CF mice do not recover efficiently, and, as a consequence of their chronic inflammation, CF mice are more susceptible to morphological changes and lung remodeling. This study shows that chronic inflammation alone contributes significantly to aspects of CF lung pathology.


Subject(s)
Cystic Fibrosis/pathology , Lipopolysaccharides/pharmacology , Lung/pathology , Pneumonia/immunology , Airway Remodeling , Animals , Chemokine CXCL10/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Lung/immunology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Knockout , Pneumonia/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
14.
Nat Commun ; 6: 6221, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25665524

ABSTRACT

In cystic fibrosis (CF) patients, hyper-inflammation is a key factor in lung destruction and disease morbidity. We have previously demonstrated that macrophages drive the lung hyper-inflammatory response to LPS in CF mice, because of reduced levels of the scaffold protein CAV1 with subsequent uncontrolled TLR4 signalling. Here we show that reduced CAV1 and, consequently, increased TLR4 signalling, in human and murine CF macrophages and murine CF lungs, is caused by high microRNA-199a-5p levels, which are PI3K/AKT-dependent. Downregulation of microRNA-199a-5p or increased AKT signalling restores CAV1 expression and reduces hyper-inflammation in CF macrophages. Importantly, the FDA-approved drug celecoxib re-establishes the AKT/miR-199a-5p/CAV1 axis in CF macrophages, and ameliorates lung hyper-inflammation in Cftr-deficient mice. Thus, we identify the AKT/miR-199a-5p/CAV1 pathway as a regulator of innate immunity, which is dysfunctional in CF macrophages contributing to lung hyper-inflammation. In addition, we found that this pathway can be targeted by celecoxib.


Subject(s)
Caveolin 1/metabolism , Cystic Fibrosis/pathology , Inflammation/pathology , Lung/pathology , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Celecoxib/pharmacology , Cystic Fibrosis/enzymology , Humans , Lung/enzymology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
15.
Blood ; 123(19): 3027-36, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24574460

ABSTRACT

Serum response factor (SRF) is a ubiquitously expressed transcription factor and master regulator of the actin cytoskeleton. We have previously shown that SRF is essential for megakaryocyte maturation and platelet formation and function. Here we elucidate the role of SRF in neutrophils, the primary defense against infections. To study the effect of SRF loss in neutrophils, we crossed Srf(fl/fl) mice with select Cre-expressing mice and studied neutrophil function in vitro and in vivo. Despite normal neutrophil numbers, neutrophil function is severely impaired in Srf knockout (KO) neutrophils. Srf KO neutrophils fail to polymerize globular actin to filamentous actin in response to N-formyl-methionine-leucine-phenylalanine, resulting in significantly disrupted cytoskeletal remodeling. Srf KO neutrophils fail to migrate to sites of inflammation in vivo and along chemokine gradients in vitro. Polarization in response to cytokine stimuli is absent and Srf KO neutrophils show markedly reduced adhesion. Integrins play an essential role in cellular adhesion, and although integrin expression levels are maintained with loss of SRF, integrin activation and trafficking are disrupted. Migration and cellular adhesion are essential for normal cell function, but also for malignant processes such as metastasis, underscoring an essential function for SRF and its pathway in health and disease.


Subject(s)
Cell Movement/genetics , Inflammation/genetics , Neutrophils/metabolism , Serum Response Factor/genetics , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Blotting, Western , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Movement/physiology , Chemokines/metabolism , Gene Expression/drug effects , Inflammation/physiopathology , Integrins/genetics , Integrins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/pathology , Polymerization/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Factor/deficiency , Serum Response Factor/physiology , Signal Transduction/genetics
16.
Stem Cells ; 31(12): 2759-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23681901

ABSTRACT

The view that adult stem cells are lineage restricted has been challenged by numerous reports of bone marrow (BM)-derived cells giving rise to epithelial cells. Previously, we demonstrated that nonhematopoietic BM cells are the primary source of BM-derived lung epithelial cells. Here, we tested the hypothesis that very small embryonic like cells (VSELs) are responsible for this engraftment. We directly compared the level of BM-derived epithelial cells after transplantation of VSELs, hematopoietic stem/progenitor cells, or other nonhematopoietic cells. VSELs clearly had the highest rate of forming epithelial cells in the lung. By transplanting VSELs from donor mice expressing H2B-GFP under a type 2 pneumocyte-specific promoter, we demonstrate that this engraftment occurs by differentiation and not fusion. This is the first report of VSELs differentiating into an endodermal lineage in vivo, thereby potentially crossing germ layer lineages. Our data suggest that Oct4+ VSELs in the adult BM exhibit broad differentiation potential.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Transplantation/methods , Embryonic Stem Cells/cytology , Lung/cytology , Animals , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Guided Tissue Regeneration , Guinea Pigs , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Immunol ; 190(10): 5196-206, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23606537

ABSTRACT

We have previously reported that TLR4 signaling is increased in LPS-stimulated cystic fibrosis (CF) macrophages (MΦs), contributing to the robust production of proinflammatory cytokines. The heme oxygenase-1 (HO-1)/CO pathway modulates cellular redox status, inflammatory responses, and cell survival. The HO-1 enzyme, together with the scaffold protein caveolin 1 (CAV-1), also acts as a negative regulator of TLR4 signaling in MΦs. In this study, we demonstrate that in LPS-challenged CF MΦs, HO-1 does not compartmentalize normally to the cell surface and instead accumulates intracellularly. The abnormal HO-1 localization in CF MΦs in response to LPS is due to decreased CAV-1 expression, which is controlled by the cellular oxidative state, and is required for HO-1 delivery to the cell surface. Overexpression of HO-1 or stimulating the pathway with CO-releasing molecules enhances CAV-1 expression in CF MΦs, suggesting a positive-feed forward loop between HO-1/CO induction and CAV-1 expression. These manipulations re-established HO-1 and CAV-1 cell surface localization in CF MΦs. Consistent with restoration of HO-1/CAV-1-negative regulation of TLR4 signaling, genetic or pharmacological (CO-releasing molecule 2) induced enhancement of this pathway decreased the inflammatory response of CF MΦs and CF mice treated with LPS. In conclusion, our results demonstrate that the counterregulatory HO-1/CO pathway, which is critical in balancing and limiting the inflammatory response, is defective in CF MΦs through a CAV-1-dependent mechanism, exacerbating the CF MΦ response to LPS. This pathway could be a potential target for therapeutic intervention for CF lung disease.


Subject(s)
Caveolin 1/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Heme Oxygenase-1/metabolism , Macrophages/immunology , Macrophages/metabolism , Adolescent , Adult , Animals , Caveolin 1/biosynthesis , Cells, Cultured , Child , Child, Preschool , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Female , Heme Oxygenase-1/biosynthesis , Humans , Inflammation/immunology , Lipopolysaccharides/immunology , Lung Diseases/immunology , Lung Diseases/metabolism , Male , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nasal Polyps , Reactive Oxygen Species/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Young Adult
18.
Stem Cells ; 30(3): 491-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22162244

ABSTRACT

Previous studies have demonstrated that bone marrow (BM)-derived cells differentiate into nonhematopoietic cells of multiple tissues. To date, it remains unknown which population(s) of BM cells are primarily responsible for this engraftment. To test the hypothesis that nonhematopoietic stem cells in the BM are the primary source of marrow-derived lung epithelial cells, either wild-type hematopoietic or nonhematopoietic BM cells were transplanted into irradiated surfactant-protein-C (SPC)-null mice. Donor-derived, SPC-positive type 2 pneumocytes were predominantly detected in the lungs of mice receiving purified nonhematopoietic cells and were absent from mice receiving purified hematopoietic stem and progenitor cells. We conclude that cells contained in the nonhematopoietic fraction of the BM are the primary source of marrow-derived lung epithelial cells. These nonhematopoietic cells may represent a primitive stem cell population residing in adult BM.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation , Epithelial Cells/metabolism , Lung/cytology , Animals , Bacterial Proteins/biosynthesis , Bone Marrow Cells/metabolism , Cell Separation , Gene Expression , Luminescent Proteins/biosynthesis , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Recombinant Proteins/biosynthesis , Single-Cell Analysis
19.
J Immunol ; 186(12): 6990-8, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21593379

ABSTRACT

Morbidity and mortality in cystic fibrosis (CF) are due not only to abnormal epithelial cell function, but also to an abnormal immune response. We have shown previously that macrophages lacking CF transmembrane conductance regulator (CFTR), the gene mutated in CF, contribute significantly to the hyperinflammatory response observed in CF. In this study, we show that lack of functional CFTR in murine macrophages causes abnormal TLR4 subcellular localization. Upon LPS stimulation, CFTR macrophages have prolonged TLR4 retention in the early endosome and reduced translocation into the lysosomal compartment. This abnormal TLR4 trafficking leads to increased LPS-induced activation of the NF-κB, MAPK, and IFN regulatory factor-3 pathways and decreased TLR4 degradation, which affects downregulation of the proinflammatory state. In addition to primary murine cells, mononuclear cells isolated from CF patients demonstrate similar defects in response to LPS. Moreover, specific inhibition of CFTR function induces abnormal TLR4 trafficking and enhances the inflammatory response of wild-type murine cells to LPS. Thus, functional CFTR in macrophages influences TLR4 spatial and temporal localization and perturbs LPS-mediated signaling in both murine CF models and patients with CF.


Subject(s)
Cystic Fibrosis/immunology , Inflammation/immunology , Toll-Like Receptor 4/metabolism , Animals , Cystic Fibrosis/pathology , Humans , Mice , Protein Transport/immunology
20.
Behav Genet ; 41(1): 58-66, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21042874

ABSTRACT

Reading disability (RD) or dyslexia is a common neurogenetic disorder. Two genes, KIAA0319 and DCDC2, have been identified by association studies of the DYX2 locus on 6p21.3. We previously identified a 2445 bp deletion, and a compound STR within the deleted region (BV677278), in intron 2 of DCDC2. The deletion and several alleles of the STR are strongly associated with RD (P = 0.00002). In this study we investigated whether BV677278 is a regulatory region for DCDC2 by electrophoretic mobility shift and luciferase reporter assays. We show that oligonucleotide probes from the STR bind nuclear protein from human brain, and that alleles of the STR have a range of DCDC2-specific enhancer activities. Five alleles displayed strong enhancer activity and increased gene expression, while allele 1 showed no enhancer activity. These studies suggest that the association of BV677278 with RD reflects a role as a modifier of DCDC2 expression.


Subject(s)
Alleles , Dyslexia/genetics , Gene Expression/genetics , Genetic Variation/genetics , Microtubule-Associated Proteins/genetics , Brain/metabolism , Cell Line , Chromosome Deletion , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , Genetic Linkage , Humans , Introns/genetics , Microsatellite Repeats/genetics , Oligonucleotide Probes , Regulatory Sequences, Nucleic Acid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...