Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Thorax ; 68(1): 82-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23093652

ABSTRACT

BACKGROUND: Overexpression of the transforming growth factor ß family signalling molecule smad2 in the airway epithelium provokes enhanced allergen-induced airway remodelling in mice, concomitant with elevated levels of interleukin (IL)-25. OBJECTIVE: We investigated whether IL-25 plays an active role in driving this airway remodelling. METHODS: Anti-IL-25 antibody was given to mice exposed to either inhaled house dust mite (HDM) alone, or in conjunction with an adenoviral smad2 vector which promotes an enhanced remodelling phenotype. RESULTS: Blocking IL-25 in allergen-exposed mice resulted in a moderate reduction in pulmonary eosinophilia and levels of T helper type 2 associated cytokines, IL-5 and IL-13. In addition, IL-25 neutralisation abrogated peribronchial collagen deposition, airway smooth muscle hyperplasia and airway hyperreactivity in control mice exposed to HDM and smad2-overexpressing mice. IL-25 was shown to act directly on human fibroblasts to induce collagen secretion. Recruitment of endothelial progenitor cells to the lung and subsequent neovascularisation was also IL-25 dependent, demonstrating a direct role for IL-25 during angiogenesis in vivo. Moreover, the secretion of innate epithelial derived cytokines IL-33 and thymic stromal lymphopoietin (TSLP) was completely ablated. CONCLUSIONS: In addition to modulating acute inflammation, we now demonstrate a role for IL-25 in orchestrating airway remodelling. IL-25 also drives IL-33 and TSLP production in the lung. These data delineate a wider role for IL-25 in mediating structural changes to the lung following allergen exposure and implicate IL-25 as a novel therapeutic target for the treatment of airway remodelling in asthma.


Subject(s)
Airway Remodeling/immunology , Bronchial Hyperreactivity/immunology , Interleukins/immunology , Pyroglyphidae/immunology , Airway Remodeling/genetics , Animals , Asthma/immunology , Asthma/metabolism , Biopsy, Needle , Bronchial Hyperreactivity/metabolism , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Regulation/immunology , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Random Allocation , Sensitivity and Specificity , Smad2 Protein/immunology
2.
FASEB J ; 26(1): 387-96, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21957127

ABSTRACT

Under homeostatic conditions, a proportion of senescent CXCR4(hi) neutrophils home from the circulation back to the bone marrow, where they are phagocytosed by bone marrow macrophages. In this study, we have identified an unexpected role for the anti-inflammatory molecule annexin A1 (AnxA1) as a critical regulator of this process. We first observed that AnxA1(-/-) mice have significantly increased neutrophil numbers in their bone marrow while having normal levels of GM and G colony-forming units, monocytes, and macrophages. Although AnxA1(-/-) mice have more neutrophils in the bone marrow, a greater proportion of these cells are senescent, as determined by their higher levels of CXCR4 expression and annexin V binding. Consequently, bone marrow neutrophils from AnxA1(-/-) mice exhibit a reduced migratory capacity in vitro. Studies conducted in vitro also show that expression of AnxA1 is required for bone marrow macrophages, but not peritoneal macrophages, to phagocytose apoptotic neutrophils. Moreover, in vivo experiments indicate a defect in clearance of wild-type neutrophils in the bone marrow of AnxA1(-/-) mice. Thus, we conclude that expression of AnxA1 by resident macrophages is a critical determinant for neutrophil clearance in the bone marrow.


Subject(s)
Annexin A1/immunology , Bone Marrow/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Animals , Annexin A1/genetics , Annexin A1/metabolism , Bone Marrow/metabolism , Cellular Senescence/immunology , Chemotaxis/immunology , Gene Expression/immunology , Homeostasis/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Phagocytosis/immunology , Receptors, CXCR4/metabolism
3.
J Allergy Clin Immunol ; 129(4): 1000-10.e3, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22277204

ABSTRACT

BACKGROUND: IL-9-secreting (T(H)9) T cells are thought to represent a distinct T-cell subset. However, evidence for their functionality in disease is uncertain. OBJECTIVE: To define a functional phenotype for T(H)9-driven pathology in vivo. METHODS: We used fluorescence-activated cell sorting to identify circulating T(H)9 cells in atopic and nonatopic subjects. In mice we utilized a model of allergic airways disease induced by house dust mite to determine T(H)9 cell function in vivo and the role of activin A in T(H)9 generation. RESULTS: Allergic patients have elevated T(H)9 cell numbers in comparison to nonatopic donors, which correlates with elevated IgE levels. In a murine model, allergen challenge with house dust mite leads to rapid T(H)9 differentiation and proliferation, with much faster kinetics than for T(H)2 cell differentiation, resulting in the specific recruitment and activation of mast cells. The TGF-ß superfamily member activin A replicates the function of TGF-ß1 in driving the in vitro generation of T(H)9 cells. Importantly, the in vivo inhibition of T(H)9 differentiation induced by allergen was achieved only when activin A and TGF-ß were blocked in conjunction but not alone, resulting in reduced airway hyperreactivity and collagen deposition. Conversely, adoptive transfer of T(H)9 cells results in enhanced pathology. CONCLUSION: Our data identify a distinct functional role for T(H)9 cells and outline a novel pathway for their generation in vitro and in vivo. Functionally, T(H)9 cells promote allergic responses resulting in enhanced pathology mediated by the specific recruitment and activation of mast cells in the lungs.


Subject(s)
Activins/pharmacology , Respiratory Hypersensitivity/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transforming Growth Factor beta/pharmacology , Activins/antagonists & inhibitors , Adoptive Transfer , Allergens/immunology , Animals , Cell Differentiation , Humans , Immunophenotyping , Lung/immunology , Lung/pathology , Lymphocyte Activation/immunology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Phenotype , Pyroglyphidae/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , T-Lymphocytes, Helper-Inducer/cytology , Transforming Growth Factor beta/antagonists & inhibitors
4.
Am J Respir Crit Care Med ; 183(7): 865-75, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-20971830

ABSTRACT

RATIONALE: IL-9 is a pleiotropic cytokine that has multiple effects on structural as well as numerous hematopoietic cells, which are central to the pathogenesis of asthma. OBJECTIVES: The contribution of IL-9 to asthma pathogenesis has thus far been unclear, due to conflicting reports in the literature. These earlier studies focused on the role of IL-9 in acute inflammatory models; here we have investigated the effects of IL-9 blockade during chronic allergic inflammation. METHODS: Mice were exposed to either prolonged ovalbumin or house dust mite allergen challenge to induce chronic inflammation and airway remodeling. MEASUREMENTS AND MAIN RESULTS: We found that IL-9 governs allergen-induced mast cell (MC) numbers in the lung and has pronounced effects on chronic allergic inflammation. Anti-IL-9 antibody-treated mice were protected from airway remodeling with a concomitant reduction in mature MC numbers and activation, in addition to decreased expression of the profibrotic mediators transforming growth factor-ß1, vascular endothelial growth factor, and fibroblast growth factor-2 in the lung. Airway remodeling was associated with impaired lung function in the peripheral airways and this was reversed by IL-9 neutralization. In human asthmatic lung tissue, we identified MCs as the main IL-9 receptor expressing population and found them to be sources of vascular endothelial growth factor and fibroblast growth factor-2. CONCLUSIONS: Our data suggest an important role for an IL-9-MC axis in the pathology associated with chronic asthma and demonstrate that an impact on this axis could lead to a reduction in chronic inflammation and improved lung function in patients with asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Bronchoalveolar Lavage Fluid/cytology , Interleukin-9/immunology , Lung/immunology , Lung/pathology , Mast Cells/immunology , Allergens/administration & dosage , Analysis of Variance , Animals , Asthma/metabolism , Biomarkers/metabolism , Biopsy, Needle , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Ovalbumin/pharmacology , RNA, Messenger/analysis , Random Allocation , Respiratory Function Tests , Statistics, Nonparametric
5.
Nat Commun ; 13(1): 2885, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35610203

ABSTRACT

Myeloid cells are central to homeostasis and immunity. Characterising in vitro myelopoiesis protocols is imperative for their use in research, immunotherapies, and understanding human myelopoiesis. Here, we generate a >470K cells molecular map of human induced pluripotent stem cells (iPSC) differentiation into macrophages. Integration with in vivo single-cell atlases shows in vitro differentiation recapitulates features of yolk sac hematopoiesis, before definitive hematopoietic stem cells (HSC) emerge. The diversity of myeloid cells generated, including mast cells and monocytes, suggests that HSC-independent hematopoiesis can produce multiple myeloid lineages. We uncover poorly described myeloid progenitors and conservation between in vivo and in vitro regulatory programs. Additionally, we develop a protocol to produce iPSC-derived dendritic cells (DC) resembling cDC2. Using CRISPR/Cas9 knock-outs, we validate the effects of key transcription factors in macrophage and DC ontogeny. This roadmap of myeloid differentiation is an important resource for investigating human fetal hematopoiesis and new therapeutic opportunities.


Subject(s)
Induced Pluripotent Stem Cells , Myelopoiesis , Cell Differentiation/genetics , Cell Lineage/genetics , Genomics , Hematopoiesis/genetics , Humans , Myelopoiesis/genetics
6.
Am J Respir Crit Care Med ; 182(2): 143-54, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20339149

ABSTRACT

RATIONALE: Airway hyperreactivity and remodeling are characteristic features of asthma. Interactions between the airway epithelium and environmental allergens are believed to be important in driving development of pathology, particularly because altered epithelial gene expression is common in individuals with asthma. OBJECTIVES: To investigate the interactions between a modified airway epithelium and a common aeroallergen in vivo. METHODS: We used an adenoviral vector to generate mice overexpressing the transforming growth factor-beta signaling molecule, Smad2, in the airway epithelium and exposed them to house dust mite (HDM) extract intranasally. MEASUREMENTS AND MAIN RESULTS: Smad2 overexpression resulted in enhanced airway hyperreactivity after allergen challenge concomitant with changes in airway remodeling. Subepithelial collagen deposition was increased and smooth muscle hyperplasia was evident resulting in thickening of the airway smooth muscle layer. However, there was no increase in airway inflammation in mice given the Smad2 vector compared with the control vector. Enhanced airway hyperreactivity and remodeling did not correlate with elevated levels of Th2 cytokines, such as IL-13 or IL-4. However, mice overexpressing Smad2 in the airway epithelium showed significantly enhanced levels of IL-25 and activin A after HDM exposure. Blocking activin A with a neutralizing antibody prevented the increase in lung IL-25 and inhibited subsequent collagen deposition and also the enhanced airway hyperreactivity observed in the Smad2 overexpressing HDM-exposed mice. CONCLUSIONS: Epithelial overexpression of Smad2 can specifically alter airway hyperreactivity and remodeling in response to an aeroallergen. Moreover, we have identified novel roles for IL-25 and activin A in driving airway hyperreactivity and remodeling.


Subject(s)
Activins/metabolism , Airway Remodeling/immunology , Bronchial Hyperreactivity/immunology , Interleukin-17/metabolism , Pyroglyphidae/immunology , Smad2 Protein/genetics , Activins/antagonists & inhibitors , Adenoviridae , Allergens/adverse effects , Animals , Antibodies/pharmacology , Collagen/biosynthesis , Dermatophagoides pteronyssinus , Disease Models, Animal , Female , Gene Expression , Goblet Cells/metabolism , Hyperplasia , Immunohistochemistry , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Muscle, Smooth/pathology , Respiratory Mucosa/metabolism
7.
Stem Cells ; 27(12): 3074-81, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19785013

ABSTRACT

Airway remodeling is a central feature of asthma and includes the formation of new peribronchial blood vessels, which is termed angiogenesis. In a number of disease models, bone marrow-derived endothelial progenitor cells (EPCs) have been shown to contribute to the angiogenic response. In this study we set out to determine whether EPCs were recruited into the lungs in a model of allergic airways disease and to identify the factors regulating EPC trafficking in this model. We observed a significant increase in the number of peribronchial blood vessels at day 24, during the acute inflammatory phase of the model. This angiogenic response was associated with an increase in the quantity of EPCs recoverable from the lung. These EPCs formed colonies after 21 days in culture and were shown to express CD31, von Willebrand factor, and vascular endothelial growth factor (VEGF) receptor 2, but were negative for CD45 and CD14. The influx in EPCs was associated with a significant increase in the proangiogenic factors VEGF-A and the CXCR2 ligands, CXCL1 and CXCL2. However, we show directly that, while the CXCL1 and CXCL2 chemokines can recruit EPCs into the lungs of allergen-sensitized mice, VEGF-A was ineffective in this respect. Further, the blockade of CXCR2 significantly reduced EPC numbers in the lungs after allergen exposure and led to a decrease in the numbers of peribronchial blood vessels after allergen challenge with no effect on inflammation. The data presented here provide in vivo evidence that CXCR2 is critical for both EPC recruitment and the angiogenic response in this model of allergic inflammation of the airways.


Subject(s)
Airway Remodeling , Cell Movement , Endothelial Cells/immunology , Hypersensitivity/immunology , Pneumonia/immunology , Receptors, Interleukin-8B/immunology , Stem Cells/immunology , Animals , Antibodies/immunology , Antibodies/therapeutic use , Cells, Cultured , Chickens , Endothelial Cells/cytology , Female , Hypersensitivity/drug therapy , Hypersensitivity/pathology , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic , Ovalbumin/immunology , Pneumonia/drug therapy , Pneumonia/pathology , Receptors, Interleukin-8B/antagonists & inhibitors , Stem Cells/cytology
8.
Science ; 362(6419): 1161-1164, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30409806

ABSTRACT

We estimated the genome-wide contribution of recessive coding variation in 6040 families from the Deciphering Developmental Disorders study. The proportion of cases attributable to recessive coding variants was 3.6% in patients of European ancestry, compared with 50% explained by de novo coding mutations. It was higher (31%) in patients with Pakistani ancestry, owing to elevated autozygosity. Half of this recessive burden is attributable to known genes. We identified two genes not previously associated with recessive developmental disorders, KDM5B and EIF3F, and functionally validated them with mouse and cellular models. Our results suggest that recessive coding variants account for a small fraction of currently undiagnosed nonconsanguineous individuals, and that the role of noncoding variants, incomplete penetrance, and polygenic mechanisms need further exploration.


Subject(s)
Developmental Disabilities/genetics , Genes, Recessive , Genetic Code , Genetic Variation , Penetrance , Animals , Disease Models, Animal , Eukaryotic Initiation Factor-3/genetics , Europe , Genome-Wide Association Study , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Nuclear Proteins/genetics , Pakistan , Phylogeny , Repressor Proteins/genetics
9.
Int Immunopharmacol ; 5(5): 857-70, 2005 May.
Article in English | MEDLINE | ID: mdl-15778122

ABSTRACT

We have previously reported that, in IL-5-stimulated bone-marrow cultures, dexamethasone upregulates eosinophil differentiation and protects developing eosinophils from apoptosis induced by a variety of agents. Recently developed procedures for the isolation of hemopoietic cells from allergic murine lungs have enabled us to evaluate how these cells respond to dexamethasone in IL-5-stimulated cultures, when compared with bone-marrow-derived cells isolated from the same donors, and whether differences in response patterns were linked to apoptosis. Ovalbumin challenge of sensitized mice increased significantly the numbers of mature leukocytes as well as hemopoietic cells recovered from digested lung fragments, relative to saline-challenged, sensitized controls. Both mature eosinophils and cells capable of differentiating into eosinophils in the presence of IL-5 were present in lungs from sensitized mice 24 h after airway challenge. Dexamethasone strongly inhibited eosinophil differentiation in IL-5-stimulated cultures of lung hemopoietic cells. By contrast, dexamethasone enhanced eosinophil differentiation in cultures of allergic bone-marrow cells, in identical conditions. Hemopoietic cells from lungs and bone-marrow were respectively susceptible and resistant to induction of apoptosis by dexamethasone. The dexamethasone-sensitive step was the response to IL-5 in culture, while accumulation of IL-5 responsive cells in allergen-challenged lungs was dexamethasone-resistant. Cells from lungs and bone-marrow, cultured for 3 days with IL-5 in the absence of dexamethasone, did not respond to a subsequent exposure to dexamethasone in the presence of IL-5. These findings confirm that IL-5-responsive hemopoietic cells found in challenged, sensitized murine lungs differ from those in bone-marrow, with respect to the cellular responses induced by dexamethasone, including apoptosis.


Subject(s)
Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Hypersensitivity/immunology , Interleukin-5/pharmacology , Lung/drug effects , Lung/immunology , Animals , Apoptosis/drug effects , Bone Marrow Cells/pathology , Dexamethasone/pharmacology , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/pathology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Hypersensitivity/pathology , Immunization , In Vitro Techniques , Lung/pathology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Recombinant Proteins/pharmacology
10.
PLoS One ; 8(7): e69246, 2013.
Article in English | MEDLINE | ID: mdl-23935964

ABSTRACT

UNLABELLED: HIF-1α is a transcription factor that is activated during hypoxia and inflammation and is a key regulator of angiogenesis in vivo. During the development of asthma, peribronchial angiogenesis is induced in response to aeroallergens and is thought to be an important feature of sustained chronic allergic inflammation. Recently, elevated HIF-1α levels have been demonstrated in both the lung tissue and bronchoalveolar lavage of allergic patients, respectively. Therefore, we investigated the role of HIF-1α on the development of angiogenesis and inflammation following acute and chronic allergen exposure. Our data shows that intranasal exposure to house dust mite (HDM) increases the expression of HIF-1α in the lung, whilst reducing the expression of the HIF-1α negative regulators, PHD1 and PHD3. Blockade of HIF-1α in vivo, significantly decreased allergic inflammation and eosinophilia induced by allergen, due to a reduction in the levels of IL-5 and Eotaxin-2. Importantly, HIF-1α blockade significantly decreased levels of VEGF-A and CXCL1 in the lungs, which in turn led to a profound decrease in the recruitment of endothelial progenitor cells and a reduction of peribronchial angiogenesis. Furthermore, HDM or IL-4 treatment of primary lung macrophages resulted in significant production of both VEGF-A and CXCL1; inhibition of HIF-1α activity abrogated the production of these factors via an up-regulation of PHD1 and PHD3. These findings suggest that novel strategies to reduce the expression and activation of HIF-1α in lung macrophages may be used to attenuate allergen-induced airway inflammation and angiogenesis through the modulation of VEGF-A and CXCL1 expression. CLINICAL RELEVANCE: This study provides new insights into the role of HIF-1α in the development of peribronchial angiogenesis and inflammation in a murine model of allergic airway disease. These findings indicate that strategies to reduce activation of macrophage derived HIF-1α may be used as a target to improve asthma pathology.


Subject(s)
Airway Remodeling , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/pathology , Lung/physiopathology , Macrophages/metabolism , Macrophages/parasitology , Animals , Chemokine CCL24/metabolism , Chemokine CXCL1/metabolism , Endothelial Cells/metabolism , Eosinophilia/complications , Eosinophilia/pathology , Eosinophilia/physiopathology , Female , Hypersensitivity/complications , Hypersensitivity/parasitology , Hypersensitivity/pathology , Hypersensitivity/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Inflammation/complications , Inflammation/pathology , Inflammation/physiopathology , Lung/parasitology , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic , Pyroglyphidae , Stem Cells/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
11.
Chest ; 140(1): 205-211, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21729891

ABSTRACT

Adult bone marrow contains a number of discrete populations of progenitor cells, including endothelial, mesenchymal, and epithelial progenitor cells and fibrocytes. In the context of a range of diseases, endothelial progenitor cells have been reported to promote angiogenesis, mesenchymal stem cells are potent immunosuppressors but can also contribute directly to tissue regeneration, and fibrocytes have been shown to induce tissue fibrosis. This article provides an overview of the basic biology of these different subsets of progenitor cells, reporting their distinct phenotypes and functional activities. The differences in their secretomes are highlighted, and the relative role of cellular differentiation vs paracrine effects of progenitor cells is considered. The article reviews the literature examining the contribution of progenitor cells to the pathogenesis of respiratory disease, and discusses recent studies using bone marrow progenitor cells as stem cell therapies in the context of pulmonary hypertension, COPD, and asthma.


Subject(s)
Bone Marrow Transplantation/methods , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Respiratory Tract Diseases/surgery , Stem Cell Transplantation/methods , Humans
12.
J Pharmacol Toxicol Methods ; 61(2): 113-21, 2010.
Article in English | MEDLINE | ID: mdl-20139021

ABSTRACT

INTRODUCTION: The molecular mechanisms that control the mobilization of specific stem cell subsets from the bone marrow are currently being intensely investigated. It is anticipated that boosting the mobilization of these stem cells via pharmacological intervention will not only produce more effective strategies for bone marrow transplant patients, but also provide novel therapeutic approaches for tissue regeneration. METHODS: Measurement of stem cell mobilization by sampling peripheral blood is problematic because it is technically difficult to accurately determine absolute numbers of rare progenitor cells by blood sampling. Furthermore a rise in progenitors may be caused by release of stem cells from tissues other than the bone marrow (e.g. spleen and adipose), or indeed an inhibition of stem cell homing back to the bone marrow or other tissues. Finally it is not possible to distinguish whether the pharmacological agent is acting directly at the level of the bone marrow or mobilizing progenitors by a distinct indirect mechanism. To resolve these problems, we have developed a technique that allows perfusion of the vasculature of the hind limb bone marrow in situ in mice. In this system, the femoral artery and vein are cannulated in situ such that the femur and tibia bone marrow are perfused in isolation under anaesthesia. As such, pharmacological agents can be administered directly into the bone marrow vasculature. Mobilized cells are then collected via the femoral vein and colony assays performed in defined growth media to allow identification of haematopoietic, endothelial, and mesenchymal progenitor cells. We have used this system to determine the ability of a CXCR4 antagonist to mobilize these distinct types of progenitor cells from the bone marrow of mice pre-conditioned with either G-CSF or VEGF. RESULTS AND CONCLUSION: This isolated hind limb perfusion system has allowed comparisons to be made between cytokines (G-CSF and VEGF) that act chronically, either alone or in combination with agents that act acutely on the bone marrow (CXCR4 antagonist) on their ability to directly mobilize specific populations of stem cells. Data obtained therefore gives a more accurate understanding of the efficacy of different mobilizing strategies compared to peripheral blood analysis.


Subject(s)
Bone Marrow Cells/physiology , Hematopoietic Stem Cell Mobilization/methods , Stem Cells/physiology , Animals , Bone Marrow Cells/drug effects , Cell Count , Cytological Techniques/standards , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cell Mobilization/standards , Hindlimb/blood supply , Hindlimb/physiology , Humans , Mice , Receptors, CXCR4/antagonists & inhibitors , Regional Blood Flow/physiology , Stem Cells/drug effects , Vascular Endothelial Growth Factor A/metabolism
13.
Cell Stem Cell ; 4(1): 62-72, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19128793

ABSTRACT

G-CSF stimulates mobilization of hematopoietic progenitor cells (HPCs) from bone marrow by disrupting the CXCR4/SDF-1alpha retention axis. We show here that distinct factors and mechanisms regulate the mobilization of endothelial (EPCs) and stromal progenitor cells (SPCs). Pretreatment of mice with VEGF did not disrupt the CXCR4/SDF-1alpha chemokine axis but stimulated entry of HPCs into the cell cycle via VEGFR1, reducing their migratory capacity in vitro and suppressing their mobilization in vivo. In contrast, VEGF pretreatment enhanced EPC mobilization via VEGFR2 in response to CXCR4 antagonism. Furthermore, SPC mobilization was detected when the CXCR4 antagonist was administered to mice pretreated with VEGF, but not G-CSF. Thus, differential mobilization of progenitor cell subsets is dependent upon the cytokine milieu that regulates cell retention and proliferation. These findings may inform studies investigating mechanisms that regulate progenitor cell recruitment in disease and can be exploited to provide efficacious stem cell therapy for tissue regeneration.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Animals , Benzylamines , Bone Marrow Cells/drug effects , Cell Cycle/drug effects , Cell Shape/drug effects , Chemokine CXCL12/metabolism , Chemotaxis/drug effects , Cyclams , Cytokines/administration & dosage , Cytokines/pharmacology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Heterocyclic Compounds/pharmacology , Mice , Mice, Inbred BALB C , Neutrophils/cytology , Neutrophils/drug effects , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Stromal Cells/cytology , Stromal Cells/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors
14.
Nitric Oxide ; 11(2): 184-93, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15491851

ABSTRACT

Apoptosis, involving both CD95/CD95L interactions and their modulation by nitric oxide (NO), is central to regulation of mature eosinophil numbers. However, its role in regulating eosinophil production from bone-marrow precursors is unknown. We examined the effects of prostaglandin E2 (PGE2) and dexamethasone on eosinophil differentiation and survival in murine bone-marrow cultures, and their relationship to: NO production as well as CD95/CD95L-dependent apoptosis. Bone-marrow cultures were established with IL-5, alone or in association with PGE2, dexamethasone or both. PGE2 (10(-7)M) inhibited eosinophil differentiation by selectively inducing apoptosis in developing eosinophils. Dexamethasone (10(-7)M) protected developing eosinophils from PGE2-induced apoptosis. Since dexamethasone prevents induction of nitric oxide synthase (NOS), we evaluated the role of NO in the effects of both PGE2 and dexamethasone. NO donors (SNAP and SNP) down-modulated eosinophil precursor responses to IL-5. SNAP induced apoptosis through a dexamethasone-resistant mechanism. The NOS inhibitors, Nomega-nitro-L-arginine and aminoguanidine, blocked the effects of PGE2 on developing eosinophils. PGE2 was ineffective in bone-marrow from knockout mice lacking inducible NOS. PGE2 up-regulated CD95 and CD95L expression in developing eosinophils. Neither PGE2 nor SNAP were effective in cultures from CD95L-deficient gld mice. These data suggest that PGE2 induces apoptosis in developing eosinophils through inducible NOS, leading to NO-dependent activation of the CD95L/CD95 pathway, while dexamethasone antagonizes the effects of PGE2 on the same targets.


Subject(s)
Dexamethasone/pharmacology , Dinoprostone/pharmacology , Eosinophils/cytology , Eosinophils/metabolism , Animals , Apoptosis/drug effects , Bone Marrow Cells , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Antagonism , Eosinophils/drug effects , Fas Ligand Protein , Female , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL