Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Virol ; 97(4): e0010223, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37022164

ABSTRACT

Whether and how a local virus infection affects the hematopoietic system in the bone marrow is largely unknown, unlike with systemic infection. In this study, we showed that influenza A virus (IAV) infection leads to demand-adapted monopoiesis in the bone marrow. The beta interferon (IFN-ß) promoter stimulator 1 (IPS-1)-type I IFN-IFN-α receptor 1 (IFNAR1) axis-mediated signaling was found to induce the emergency expansion of the granulocyte-monocyte progenitor (GMP) population and upregulate the expression of the macrophage colony-stimulating factor receptor (M-CSFR) on bipotent GMPs and monocyte progenitors via the signal transducer and activator of transcription 1 (STAT1), leading to a scaled-back proportion of granulocyte progenitors. To further address the influence of demand-adapted monopoiesis on IAV-induced secondary bacterial infection, IAV-infected wild-type (WT) and Stat1-/- mice were challenged with Streptococcus pneumoniae. Compared with WT mice, Stat1-/- mice did not demonstrate demand-adapted monopoiesis, had more infiltrating granulocytes, and were able to effectively eliminate the bacterial infection. IMPORTANCE Our findings show that influenza A virus infection induces type I interferon (IFN)-mediated emergency hematopoiesis to expand the GMP population in the bone marrow. The type I IFN-STAT1 axis was identified as being involved in mediating the viral-infection-driven demand-adapted monopoiesis by upregulating M-CSFR expression in the GMP population. As secondary bacterial infections often manifest during a viral infection and can lead to severe or even fatal clinical complications, we further assessed the impact of the observed monopoiesis on bacterial clearance. Our results suggest that the resulting decrease in the proportion of granulocytes may play a role in diminishing the IAV-infected host's ability to effectively clear secondary bacterial infection. Our findings not only provide a more complete picture of the modulatory functions of type I IFN but also highlight the need for a more comprehensive understanding of potential changes in hematopoiesis during local infections to better inform clinical interventions.


Subject(s)
Interferon Type I , Orthomyxoviridae Infections , Receptor, Macrophage Colony-Stimulating Factor , STAT1 Transcription Factor , Up-Regulation , Animals , Humans , Mice , Influenza A virus/immunology , Interferon Type I/immunology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Orthomyxoviridae Infections/immunology , Hematopoiesis/immunology , Granulocyte-Macrophage Progenitor Cells/immunology , Streptococcus pneumoniae/immunology , Pneumococcal Infections/immunology
2.
Elife ; 102021 04 08.
Article in English | MEDLINE | ID: mdl-33830019

ABSTRACT

Innate immune cellular effectors are actively consumed during systemic inflammation, but the systemic traffic and the mechanisms that support their replenishment remain unknown. Here, we demonstrate that acute systemic inflammation induces the emergent activation of a previously unrecognized system of rapid migration of granulocyte-macrophage progenitors and committed macrophage-dendritic progenitors, but not other progenitors or stem cells, from bone marrow (BM) to regional lymphatic capillaries. The progenitor traffic to the systemic lymphatic circulation is mediated by Ccl19/Ccr7 and is NF-κB independent, Traf6/IκB-kinase/SNAP23 activation dependent, and is responsible for the secretion of pre-stored Ccl19 by a subpopulation of CD205+/CD172a+ conventional dendritic cells type 2 and upregulation of BM myeloid progenitor Ccr7 signaling. Mature myeloid Traf6 signaling is anti-inflammatory and necessary for lymph node myeloid cell development. This report unveils the existence and the mechanistic basis of a very early direct traffic of myeloid progenitors from BM to lymphatics during inflammation.


When the body becomes infected with disease-causing pathogens, such as bacteria, the immune system activates various mechanisms which help to fight off the infection. One of the immune system's first lines of defense is to launch an inflammatory response that helps remove the pathogen and recruit other immune cells. However, this response can become overactivated, leading to severe inflammatory conditions that damage healthy cells and tissues. A second group of cells counteract this over inflammation and are different to the ones involved in the early inflammatory response. Both types of cells ­ inflammatory and anti-inflammatory ­ develop from committed progenitors, which, unlike stem cells, are already destined to become a certain type of cell. These committed progenitors reside in the bone marrow and then rapidly travel to secondary lymphoid organs, such as the lymph nodes, where they mature into functioning immune cells. During this journey, committed progenitors pass from the bone marrow to the lymphatic vessels that connect up the different secondary lymphoid organs, and then spread to all tissues in the body. Yet, it is not fully understood what exact route these cells take and what guides them towards these lymphatic tissues during inflammation. To investigate this, Serrano-Lopez, Hegde et al. used a combination of techniques to examine the migration of progenitor cells in mice that had been treated with lethal doses of a bacterial product that triggers inflammation. This revealed that as early as one to three hours after the onset of infection, progenitor cells were already starting to travel from the bone marrow towards lymphatic vessels. Serrano-Lopez, Hegde et al. found that a chemical released by an "alarm" immune cell already residing in secondary lymphoid organs attracted these progenitor cells towards the lymphatic tissue. Further experiments showed that the progenitor cells travelling to secondary lymphoid organs were already activated by bacterial products. They then follow the chemical released by alarm immune cells ready to respond to the immune challenge and suppress inflammation. These committed progenitors were also found in the inflamed lymph nodes of patients. These findings suggest this rapid circulation of progenitors is a mechanism of defense that contributes to the fight against severe inflammation. Altering how these cells migrate from the bone marrow to secondary lymphoid organs could provide a more effective treatment for inflammatory conditions and severe infections. However, these approaches would need to be tested further in the laboratory and in clinical trials.


Subject(s)
Bone Marrow/metabolism , Cell Movement , Granulocyte-Macrophage Progenitor Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Lymphadenopathy/metabolism , Lymphatic System/metabolism , Myeloid Progenitor Cells/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Cell Lineage , Cells, Cultured , Child , Child, Preschool , Disease Models, Animal , Female , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Lymphadenopathy/immunology , Lymphadenopathy/pathology , Lymphatic System/immunology , Lymphatic System/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Phenotype , Signal Transduction , Time Factors , Young Adult
3.
Front Immunol ; 9: 2685, 2018.
Article in English | MEDLINE | ID: mdl-30510555

ABSTRACT

Innate immune memory describes the functional reprogramming of innate immune cells after pathogen contact, leading to either a boosted (trained immunity) or a diminished (immune tolerance) response to a secondary stimulus. Immune tolerance or "sepsis-induced immunosuppression" is a typical hallmark of patients after sepsis survival, characterized by hypo-responsiveness of the host's immune system. This condition renders the host vulnerable for a persisting infection or the occurrence of secondary, often opportunistic infections, along with an increased mortality rate. The mechanisms involved in the maintenance of this long-lasting condition are not examined yet. Polymicrobial abdominal sepsis was induced in 12 week old male C57BL/6 mice by cecal ligation and puncture. Mice were euthanized 3 months after insult. Immune cell composition of the spleen and whole blood, as well as stem and progenitor cells of the bone marrow, were assessed by flow cytometry. Whole blood and bone marrow monocytes were stimulated with LPS and supernatant levels of TNF and IL-6 detected by ELISA. Furthermore, naïve bone marrow monocytes were analyzed for metabolic (Seahorse technology) and transcriptomic (RNA sequencing) changes. Flow cytometric analysis revealed an increase of inflammatory monocytes and regulatory T cells in the spleen, whereby immune composition of whole blood kept unchanged. Granulocyte-monocyte progenitor cells are increased in sepsis survivors. Systemic cytokine response was unchanged after LPS challenge. In contrast, cytokine response of post-septic naïve bone marrow monocytes was increased. Metabolic analysis revealed enhanced glycolytic activity, whereas mitochondrial indices were not affected. In addition, RNA sequencing analysis of global gene expression in monocytes revealed a sustained signature of 367 differentially expressed genes. We here demonstrate that sepsis via functional reprogramming of naïve bone marrow monocytes induces a cellular state of trained immunity, which might be counteracted depending on the compartmental localization of the cell. These findings shed new light on the complex aftermath of sepsis and open up a new pathophysiological framework in need for further research.


Subject(s)
Bone Marrow Cells/immunology , Gene Expression Regulation/immunology , Granulocyte-Macrophage Progenitor Cells/immunology , Monocytes/metabolism , Sepsis/immunology , Animals , Bone Marrow Cells/pathology , Granulocyte-Macrophage Progenitor Cells/pathology , Interleukin-6/immunology , Mice , Monocytes/pathology , Sepsis/pathology , Tumor Necrosis Factor-alpha/immunology
4.
Sci Rep ; 6: 23115, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26979287

ABSTRACT

Growing evidence suggests that hematopoietic stem/progenitor cells (HSPCs), precursors of mature immune cells, may play a direct role in immunosurveillance. Early myeloid progenitors are the major components of HSPCs and they often undergo extensive expansion in stress as a result of myeloid-biased hematopoiesis. Yet, the precise function of early myeloid progenitors remains unclear. Here we show that during tumor progression, mouse granulocyte/macrophage progenitors (GMPs) but not common myeloid progenitors (CMPs) are markedly expanded within the bone marrow and blood of mice. Interestingly, both GMPs and CMPs freshly isolated from either tumor-bearing or naïve animals are capable of inhibiting polyclonal stimuli- and alloantigen-induced T cell proliferation, with tumor host-derived cells having elevated activities. Strikingly, these early myeloid progenitor cells even display much stronger suppressive capacity than the classical myeloid-derived suppressive cells. Analysis of GMPs indicates that they express iNOS and can secrete high levels of NO. Further studies unusing iNOS specific inhibitors reveal that the immunosuppression of GMPs is, to a large extent, NO-dependent. GMPs can also efficiently induce regulatory T cell development. These studies demonstrate that early myeloid progenitors can act as immunosuppressive cells. This finding provides novel insights into the functional diversity and plasticity of early myeloid progenitor cells.


Subject(s)
Carcinoma, Lewis Lung/immunology , Hematopoietic Stem Cells/immunology , Myeloid Progenitor Cells/immunology , T-Lymphocytes/immunology , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Progression , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoietic Stem Cells/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
5.
J Immunol Methods ; 425: 21-26, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26056939

ABSTRACT

Different dendritic cell (DC) subsets co-exist in humans and coordinate the immune response. Having a short life, DCs must be constantly replenished from their progenitors in the bone marrow through hematopoiesis. Identification of a DC-restricted progenitor in mouse has improved our understanding of how DC lineage diverges from myeloid and lymphoid lineages. However, identification of the DC-restricted progenitor in humans has not been possible because a system that simultaneously nurtures differentiation of human DCs, myeloid and lymphoid cells, is lacking. Here we report a cytokine and stromal cell culture that allows evaluation of CD34(+) progenitor potential to all three DC subsets as well as other myeloid and lymphoid cells, at a single cell level. Using this system, we show that human granulocyte-macrophage progenitors are heterogeneous and contain restricted progenitors to DCs.


Subject(s)
Dendritic Cells/immunology , Stem Cells/immunology , Stromal Cells/immunology , Antigens, CD34/immunology , Bone Marrow/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Cells, Cultured , Granulocyte-Macrophage Progenitor Cells/immunology , Hematopoiesis/immunology , Humans , Lymphocytes/immunology
7.
Immunity ; 39(1): 97-110, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23871207

ABSTRACT

It remains unclear whether basophils and mast cells are derived from a common progenitor. Furthermore, how basophil versus mast cell fate is specified has not been investigated. Here, we have identified a population of granulocyte-macrophage progenitors (GMPs) that were highly enriched in the capacity to differentiate into basophils and mast cells while retaining a limited capacity to differentiate into myeloid cells. We have designated these progenitor cells "pre-basophil and mast cell progenitors" (pre-BMPs). STAT5 signaling was required for the differentiation of pre-BMPs into both basophils and mast cells and was critical for inducing two downstream molecules: C/EBPα and MITF. We have identified C/EBPα as the critical basophil transcription factor for specifying basophil cell fate and MITF as the crucial transcription factor for specifying mast cell fate. C/EBPα and MITF silenced each other's transcription in a directly antagonistic fashion. Our study reveals how basophil and mast cell fate is specified.


Subject(s)
Basophils/immunology , CCAAT-Enhancer-Binding Protein-alpha/immunology , Mast Cells/immunology , Microphthalmia-Associated Transcription Factor/immunology , Animals , Basophils/cytology , Basophils/metabolism , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , HEK293 Cells , Humans , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Stem Cells/immunology , Stem Cells/metabolism
8.
J Neuroimmunol ; 259(1-2): 17-25, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23582490

ABSTRACT

Melatonin is known as an antistress and immunostimulator compound while glucocorticoids have immunosuppressive function. The mechanism of action of both the hormones on immune cells is still a question. We found that melatonin improved the effect of dexamethasone (synthetic glucocorticoid) induced immunosuppression of splenocytes and bone marrow GM-CFU along with increased production of serum IL-2, IgG and the receptor expression for melatonin and glucocorticoid in spleen that might be responsible for the proliferation of immune cells. Thus, seasonal variation in peripheral melatonin might be responsible for the improvement of immune status under different stress conditions experienced by the rodents for better survival.


Subject(s)
Dexamethasone/pharmacology , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Melatonin/pharmacology , Stress, Physiological/immunology , Animals , Animals, Outbred Strains , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Central Nervous System Depressants/immunology , Central Nervous System Depressants/metabolism , Central Nervous System Depressants/pharmacology , Cricetinae , Drug Interactions , Glucocorticoids/pharmacology , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , Hydrocortisone/blood , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunoglobulin G/blood , Interleukin-2/blood , Male , Melatonin/blood , Melatonin/immunology , Mesocricetus , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Stress, Physiological/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
9.
Exp Gerontol ; 48(2): 168-82, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23220117

ABSTRACT

Reactive oxygen species (ROS) have been proposed to play an important role in balancing the pro- and antioxidant homeostasis during aging. Melatonin has been suggested as an effective free radical scavenger that might have a role during the process of aging. We observed, that melatonin administration (25 µg/100 g body weight for 30 days) significantly augments the activity of anti-oxidative enzymes like superoxide dismutase (SOD), catalase and glutathione peroxidase (GPx) in the plasma, spleen and bone marrow (BM) of young (6 weeks), adult (30 weeks) and old aged (2.5 years) male golden hamster, Mesocricetus auratus. A sharp decline in generation of ROS was observed in peripheral blood mononuclear cells (PBMC) and splenocytes upon melatonin administration in different age group of hamsters. Reduction in the level of thiobarbituric acid-reactive substances (TBARS) and total nitrite and nitrate concentration as metabolites and indicators of nitric oxide (NO) in plasma, spleen and BM were observed along with night time (22:00 h) melatonin concentration in different age group of hamsters after administration of melatonin and compared to the control group (treated with 0.9% saline). General immune parameters like proliferation of splenocytes, PBMC and colony forming ability of GM-CFU were observed following melatonin treatment in different age group, although it was low only in aged hamsters compared to the young and adult. Our data indicates that the age related increase of oxidative load and simultaneously augments the general immunity in aged hamsters.


Subject(s)
Aging , Bone Marrow/drug effects , Free Radical Scavengers/administration & dosage , Melatonin/administration & dosage , Oxidative Stress/drug effects , Spleen/drug effects , Age Factors , Aging/immunology , Aging/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Catalase/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cricetinae , Glutathione Peroxidase/metabolism , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , Injections, Subcutaneous , Lipid Peroxidation/drug effects , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mesocricetus , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Reactive Oxygen Species/metabolism , Spleen/immunology , Spleen/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
10.
J Immunol ; 188(7): 3208-16, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22351937

ABSTRACT

The earliest thymic progenitors (ETPs) were recently shown to give rise to both lymphoid and myeloid cells. Whereas the majority of ETPs are derived from IL-7Rα-positive cells and give rise exclusively to T cells, the origin of the myeloid cells remains undefined. In this study, we show both in vitro and in vivo that IL-13Rα1(+) ETPs yield myeloid cells with no potential for maturation into T cells, whereas IL-13Rα1(-) ETPs lack myeloid potential. Moreover, transfer of lineage-negative IL-13Rα1(+) bone marrow stem cells into IL-13Rα1-deficient mice reconstituted thymic IL-13Rα1(+) myeloid ETPs. Myeloid cells or macrophages in the thymus are regarded as phagocytic cells whose function is to clear apoptotic debris generated during T cell development. However, the myeloid cells derived from IL-13Rα1(+) ETPs were found to perform Ag-presenting functions. Thus, IL-13Rα1 defines a new class of myeloid restricted ETPs yielding APCs that could contribute to development of T cells and the control of immunity and autoimmunity.


Subject(s)
Antigen-Presenting Cells/cytology , Antigens, Differentiation/analysis , Bone Marrow Cells/classification , Granulocyte-Macrophage Progenitor Cells/cytology , Interleukin-13 Receptor alpha1 Subunit/analysis , Myelopoiesis , Thymus Gland/cytology , Animals , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Bone Marrow Cells/chemistry , Cell Lineage , Cell Movement , Cells, Cultured , Female , Gene Knock-In Techniques , Granulocyte-Macrophage Progenitor Cells/chemistry , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/immunology , Interleukin-13/pharmacology , Interleukin-13 Receptor alpha1 Subunit/deficiency , Interleukin-13 Receptor alpha1 Subunit/genetics , Lymphocytes, Null/cytology , Lymphopoiesis , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Sequence Deletion , T-Lymphocytes/cytology
11.
Biol Pharm Bull ; 34(10): 1533-41, 2011.
Article in English | MEDLINE | ID: mdl-21963492

ABSTRACT

Mast-cell-development in the bone-marrow (BM) and the spleen is restrictedly controlled by stromal-cells which produce positive-regulators such as stem cell factor (SCF), and negative-regulators such as transforming growth factor-ß (TGF-ß). How the balance between positive- and negative-regulation is achieved or maintained by stromal-cells is not well understood. We intravenously injected 5-fluorouracil (5-FU) and lipopolysaccharide (LPS) into C3H/HeN mice to disrupt mast-cell-development in order to reveal mechanisms of mast-cell-regulation. 5-FU treatment induces a rapid decrease in the number of mast-cell-progenitor (colony-forming unit (CFU)-mast) cells in the BM and spleen, followed by rapid recovery of CFU-mast numbers. Expression of the SCF gene is one-fiftieth the level of that of TGF-ß during the steady-state in BM and spleen. After 5-FU treatment, SCF mRNA levels in the BM markedly increased, approaching TGF-ß mRNA levels, whereas SCF levels in the spleen showed limited oscillations whose increases paralleled those in TGF-ß levels. In contrast, LPS treatment induces a rapid decrease in CFU-mast number in the BM and a rapid increase in of CFU-mast number in the spleen. After LPS treatment, SCF mRNA levels in the BM markedly decreased, whereas SCF levels in the spleen remained unchanged. These results suggest that regulation of mast-cell-development is dominated by negative-signals in the BM and spleen during the steady-state, and, under biostress-conditions such as 5-FU and LPS treatment, the balance between positive- and negative-regulation can be changed in the BM but not in the spleen. The difference in the regulation of mast-cell-development in the BM versus the spleen probably reflects the different roles of tissue-specific stromal-cells.


Subject(s)
Cytokines/genetics , Fluorouracil/pharmacology , Granulocyte-Macrophage Progenitor Cells/immunology , Immunosuppressive Agents/pharmacology , Lipopolysaccharides/immunology , Mast Cells/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Division/drug effects , Cell Proliferation/drug effects , Colony-Forming Units Assay , Cytokines/metabolism , Drug Evaluation, Preclinical , Escherichia coli , Female , Fluorouracil/metabolism , Gene Expression/drug effects , Granulocyte-Macrophage Progenitor Cells/drug effects , Immunosuppressive Agents/metabolism , Leukocytes/drug effects , Lipopolysaccharides/metabolism , Mast Cells/metabolism , Mice , Mice, Inbred C3H , Spleen/immunology , Spleen/metabolism , Stromal Cells/physiology , Time Factors
12.
Nat Immunol ; 12(2): 144-50, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21170027

ABSTRACT

Transcription factor NF-κB and its activating kinase IKKß are associated with inflammation and are believed to be critical for innate immunity. Despite the likelihood of immune suppression, pharmacological blockade of IKKß-NF-κB has been considered as a therapeutic strategy. However, we found neutrophilia in mice with inducible deletion of IKKß (Ikkß(Δ) mice). These mice had hyperproliferative granulocyte-macrophage progenitors and pregranulocytes and a prolonged lifespan of mature neutrophils that correlated with the induction of genes encoding prosurvival molecules. Deletion of interleukin 1 receptor 1 (IL-1R1) in Ikkß(Δ) mice normalized blood cellularity and prevented neutrophil-driven inflammation. However, Ikkß(Δ)Il1r1(-/-) mice, unlike Ikkß(Δ) mice, were highly susceptible to bacterial infection, which indicated that signaling via IKKß-NF-κB or IL-1R1 can maintain antimicrobial defenses in each other's absence, whereas inactivation of both pathways severely compromises innate immunity.


Subject(s)
Bacterial Infections/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , I-kappa B Kinase/metabolism , Interleukin-1beta/metabolism , Neutrophils/metabolism , Animals , Cell Count , Cell Growth Processes/genetics , Cell Survival/genetics , Cells, Cultured , Disease Susceptibility , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/pathology , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Immunity, Innate/genetics , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Neutrophils/immunology , Neutrophils/pathology , Receptors, Interleukin-1 Type I/genetics , Up-Regulation/genetics
13.
Nutr Cancer ; 62(8): 1170-80, 2010.
Article in English | MEDLINE | ID: mdl-21058206

ABSTRACT

We studied the effects of Chlorella vulgaris (CV) on the interaction between stromal and hematopoietic stem cells in normal and Ehrlich ascites tumor (EAT)-bearing mice. Long-term bone marrow culture (LTBMC), cytokine production, spleen mononuclear cells (SMC) proliferation (SCP), colony stimulating activity (CSA), and NK cells activity were evaluated. In tumor bearers, reduced capacity of stromal cell layer to support the growth and differentiation of granulocyte-macrophage progenitor cells (CFU-GM), concomitantly to decreased numbers of total nonadherent cells in LTBMC and reduced local production of IL-6 and IL-1α, were observed. Presence of the tumor has not altered the number of stromal adherent cells. CV treatment restored the ability of stromal cells from EAT-bearing mice to produce IL-6 and IL-1α, which was consistent with increased number of nonadherent cells and higher ability to display CFU-GM in vitro. EAT growth increased SCP, serum CSA, and IL-10 production and concurrently depressed NK cell activity and the secretion of IL-2, IFN-γ, and TNF-α. Treatment of tumor-bearing mice with CV augmented CSA, SMC proliferation, NK cell activity, and the production of IL-2, IFN-γ, and TNF-α, whereas IL-10 levels where reduced. Our results suggest that CV modulates immunehematopoietic cell activity and disengages tumor-induced suppression of these responses.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Ehrlich Tumor/drug therapy , Carcinoma, Ehrlich Tumor/immunology , Chlorella vulgaris , Immunologic Factors/therapeutic use , Myelopoiesis , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Carcinoma, Ehrlich Tumor/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Colony-Stimulating Factors/blood , Colony-Stimulating Factors/metabolism , Cytokines/metabolism , Dietary Supplements , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred BALB C , Phytotherapy , Specific Pathogen-Free Organisms , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism
14.
Int Immunopharmacol ; 10(12): 1573-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20951668

ABSTRACT

In this study we demonstrated that the oral administration of ß-1,3-glucan (Imunoglucan®) protects mice from a lethal dose of Listeria monocytogenes (LM) when administered prophylactically for 10 days at the doses of 150 and 300 mg/kg, with survival rates up to 40%. These doses also prevented the myelosuppression and the splenomegaly caused by a sublethal infection with LM, due to increased numbers of granulocyte-macrophage progenitors (CFU-GM) in the bone marrow. Investigation of the production of colony-stimulating factors revealed an increased colony-stimulating activity (CSA) in the serum of infected mice pre-treated with Imunoglucan®. The treatment also restored the reduced ability of stromal cells to display myeloid progenitors in long-term bone marrow cultures (LTBMC) and up-regulated IL-6 and IL-1α production by these cells in the infected mice, which was consistent with higher number of non-adherent cells. Additional studies to investigate the levels of interferon-gamma (INF-γ) in the supernatant of splenocyte cultures demonstrated a further increase in the level of this cytokine in infected-treated mice, compared to infected controls. In all cases, no differences were observed between the responses of the two optimal biologically effective doses. In contrast, no significant changes were produced by the treatment with the 50mg/kg dose. In addition, no changes were observed in normal mice treated with the three doses used. All together our results suggest that orally given Imunoglucan® indirectly modulates immune activity and probably disengages Listeria induced suppression of these responses by inducing a higher reserve of myeloid progenitors in the bone marrow in consequence of biologically active cytokine release (CSFs, IL-1α, IL-6, and INF-γ).


Subject(s)
Adjuvants, Immunologic/therapeutic use , Hematopoiesis/drug effects , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Listeriosis/prevention & control , beta-Glucans/therapeutic use , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Culture Techniques , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Listeria monocytogenes/drug effects , Listeriosis/complications , Listeriosis/immunology , Listeriosis/microbiology , Male , Mice , Mice, Inbred BALB C , Splenomegaly/etiology , Splenomegaly/immunology , Splenomegaly/prevention & control , beta-Glucans/administration & dosage
15.
Cancer Immunol Immunother ; 59(6): 885-97, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20140432

ABSTRACT

Bone marrow myelotoxicity is a major limitation of chemotherapy. While granulocyte colony stimulating factor (G-CSF) treatment is effective, alternative approaches to support hematopoietic recovery are sought. We previously found that a beta-glucan extract from maitake mushroom Grifola frondosa (MBG) enhanced colony forming unit-granulocyte monocyte (CFU-GM) activity of mouse bone marrow and human hematopoietic progenitor cells (HPC), stimulated G-CSF production and spared HPC from doxorubicin toxicity in vitro. This investigation assessed the effects of MBG on leukocyte recovery and granulocyte/monocyte function in vivo after dose intensive paclitaxel (Ptx) in a normal mouse. After a cumulative dose of Ptx (90-120 mg/kg) given to B6D2F1mice, daily oral MBG (4 or 6 mg/kg), intravenous G-CSF (80 microg/kg) or Ptx alone were compared for effects on the dynamics of leukocyte recovery in blood, CFU-GM activity in bone marrow and spleen, and granulocyte/monocyte production of reactive oxygen species (ROS). Leukocyte counts declined less in Ptx + MBG mice compared to Ptx-alone (p = 0.024) or Ptx + G-CSF treatment (p = 0.031). Lymphocyte levels were higher after Ptx + MBG but not Ptx + G-CSF treatment compared to Ptx alone (p < 0.01). MBG increased CFU-GM activity in bone marrow and spleen (p < 0.001, p = 0.002) 2 days after Ptx. After two additional days (Ptx post-day 4), MBG restored granulocyte/monocyte ROS response to normal levels compared to Ptx-alone and increased ROS response compared to Ptx-alone or Ptx + G-CSF (p < 0.01, both). The studies indicate that oral MBG promoted maturation of HPC to become functionally active myeloid cells and enhanced peripheral blood leukocyte recovery after chemotoxic bone marrow injury.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Progenitor Cells/metabolism , Grifola , Leukocytes/metabolism , Paclitaxel/administration & dosage , beta-Glucans/administration & dosage , Animals , Bone Marrow/drug effects , Bone Marrow/pathology , Cell Line, Tumor , Dietary Carbohydrates/administration & dosage , Drug Antagonism , Drug Therapy, Combination , Granulocyte-Macrophage Progenitor Cells/drug effects , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/pathology , Leukopoiesis/drug effects , Mice , Oxidation-Reduction/drug effects , Paclitaxel/adverse effects , Reactive Oxygen Species/metabolism
16.
Blood ; 114(21): 4721-8, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19779039

ABSTRACT

Neutrophils play a vital role in the immune defense, which is evident by the severity of neutropenia causing life-threatening infections. Granulocyte macrophage-colony stimulating factor (GM-CSF) controls homeostatic and emergency development of granulocytes. However, little is known about the contribution of the downstream mediating transcription factors signal transducer and activator of transcription 5A and 5B (STAT5A/B). To elucidate the function of this pathway, we generated mice with complete deletion of both Stat5a/b genes in hematopoietic cells. In homeostasis, peripheral neutrophils were markedly decreased in these animals. Moreover, during emergency situations, such as myelosuppression, Stat5a/b-mutant mice failed to produce enhanced levels of neutrophils and were unable to respond to GM-CSF. Both the GM-CSF-permitted survival of mature neutrophils and the generation of granulocytes from granulocyte-macrophage progenitors (GMPs) were markedly reduced in Stat5a/b mutants. GMPs showed impaired colony-formation ability with reduced number and size of colonies on GM-CSF stimulation. Moreover, continuous cell fate analyses by time-lapse microscopy and single cell tracking revealed that Stat5a/b-null GMPs showed both delayed cell-cycle progression and increased cell death. Finally, transcriptome analysis indicated that STAT5A/B directs GM-CSF signaling through the regulation of proliferation and survival genes.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Leukopoiesis/physiology , Neutrophils/cytology , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Gene Expression Profiling , Gene Expression Regulation/immunology , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocytes/cytology , Granulocytes/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Oligonucleotide Array Sequence Analysis
17.
Pediatr Hematol Oncol ; 26(5): 338-44, 2009.
Article in English | MEDLINE | ID: mdl-19579080

ABSTRACT

Thalassemic children are at a high risk of graft rejection in cord blood transplantation. To investigate this possible mechanism, the authors evaluated the effect of panel reactive antibody on the growth of CD34(+) cells in vitro. On semisolid medium, CD34(+) cells derived from cord blood were incubated with thalassemic serum, and colony-forming units were counted after 10 days of culture. After incubation with serum-specific panel reactive antibody, profound decreases were found in the numbers of CFU-GM, CFU-GEMM and BFU-E compared with controls. The results indicated that serum-specific panel reactive antibody might have an apparent inhibition effect on proliferation and differentiation of cord blood CD34(+) cells.


Subject(s)
Antibodies/immunology , Antigens, CD34/immunology , Erythroid Precursor Cells/immunology , Fetal Blood/immunology , Granulocyte-Macrophage Progenitor Cells/immunology , Myeloid Progenitor Cells/immunology , beta-Thalassemia/immunology , Antibodies/blood , Apoptosis/immunology , Cell Differentiation/immunology , Cells, Cultured , Child , Erythroid Precursor Cells/cytology , Fetal Blood/cytology , Granulocyte-Macrophage Progenitor Cells/cytology , Humans , Myeloid Progenitor Cells/cytology , beta-Thalassemia/blood
18.
J Immunol ; 182(5): 2835-41, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234178

ABSTRACT

Recent work has established important roles for basophils in regulating immune responses. To exert their biological functions, basophils need to be expanded to critical numbers. However, the mechanisms underlying basophil expansion remain unclear. In this study, we established that IL-3 played an important role in the rapid and specific expansion of basophils. We found that the IL-3 complex (IL-3 plus anti-IL-3 Ab) greatly facilitated the differentiation of GMPs into basophil lineage-restricted progenitors (BaPs) but not into eosinophil lineage-restricted progenitors or mast cells in the bone marrow. We also found that the IL-3 complex treatment resulted in approximately 4-fold increase in the number of basophil/mast cell progenitors (BMCPs) in the spleen. IL-3-driven basophil expansion depended on STAT5 signaling. We showed that GMPs but not common myeloid progenitors expressed low levels of IL-3 receptor. IL-3 receptor expression was dramatically up-regulated in BaPs but not eosinophil lineage-restricted progenitors. Approximately 38% of BMCPs expressed the IL-3R alpha-chain. The up-regulated IL-3 receptor expression was not affected by IL-3 or STAT5. Our findings demonstrate that IL-3 induced specific expansion of basophils by directing GMPs to differentiate into BaPs in the bone marrow and by increasing the number of BMCPs in the spleen.


Subject(s)
Basophils/immunology , Cell Differentiation/immunology , Cell Lineage/immunology , Granulocyte Precursor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/immunology , Interleukin-3/physiology , Spleen/immunology , Up-Regulation/immunology , Animals , Basophils/cytology , Basophils/metabolism , Gene Expression Regulation/immunology , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Interleukin-3/administration & dosage , Interleukin-3/deficiency , Interleukin-3/genetics , Leukocyte Count , Mast Cells/cytology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/metabolism , Receptors, Interleukin-3/biosynthesis , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/physiology , Spleen/cytology , Spleen/metabolism , Up-Regulation/genetics
19.
Zhonghua Yi Xue Za Zhi ; 88(48): 3401-6, 2008 Dec 30.
Article in Chinese | MEDLINE | ID: mdl-19159569

ABSTRACT

OBJECTIVE: To investigate the effects of targeting elimination of the leukemic CD34+ progenitor cells by using cytotoxic T lymphocytes (CTLs) specifically against Wilms tumor gene (WT1)-derived peptide. METHODS: A 9-mer WT1 peptide (CMTWNQMNL) containing HLA-A * 0201-binding anchor motifs was synthesized. The suspended cells were collected from the culture of the peripheral blood mononuclear cells and divided into 2 groups: Group D (pure T cells) and Group C (IL2 + T cells). The dendritic cells (DCs) generated from the peripheral blood mononuclear cells of an HLA-A* 0201-positive healthy donor were repeatedly loaded with WT1 peptide so as to elicit cytotoxic T cells (CTLs) specifically for WT1 peptide, and restricted by HLA-A * 0201 (Group A). The specific lysis effects of WT1 peptide specific CTLs upon leukemic bone marrow CD34+ progenitor cells positively expressing WT1 (3 being HLA-A * 0201(+) and 3 being HLA-A*0201(-)), peripheral blood CD34+ cells from healthy persons (2 being HLA-A * 0201(+) and 1 being HLA-A * 0201(-)), and leukemia cells of the lines of NB4 (HLA-A * 0201(+)/WT1(+)), U937 (HLA-A * 0201(+)/WT1(-)), and K562 (HLA-A * 0201(-)/WT1(+)) were measured by methyl thiazolyl tetrazolium (MTT) chromatometry assay. The colony-forming unit-granulocyte macrophage (CFU-GM) forming capability of the leukemic bone marrow CD34+ progenitor cells and peripheral blood CD34+ cells from healthy persons treated with WT1 peptide specific CTLs were also determined by methylcellulose medium colony-forming unit assay. The CTLs elicited by DCs without WT1 peptide loading(Group B)and T cells cultured with IL-2 (Group C) were taken as controls. RESULTS: The CTLs specific for WT1 peptide and restricted by HLA-A * 0201 (Group A) exerted a specific lysis upon the 3 cases with HLA-A * 0201(+)/WT1(+) leukemic bone marrow CD34+ progenitor cells and NB4 leukemia cells, the specific lysis levels were 55.3% +/- 2.8%, 67.1% +/- 3.2%, 49.4% +/- 3.8% and 55.0% +/- 3.7% respectively, all significantly higher than those upon the 3 cases with HLA-A * 0201(-)/WT1(+) leukemic bone marrow CD34+ progenitor cells, normal healthy peripheral blood CD34+ cells of the healthy persons, and leukemia cell of the lines K562 and U937 (the specific lysis levels were all < 20%). The specific lysis level of Group A CTLs was significantly higher than those of Group B and Group C CTLs (both P < 0.01). The relative colony formation rates of WT1-CTLs in Group A upon the 2 cases with HLA-A * 0201(+)/WT1(+) leukemic CD34+ progenitor cells were 17.8% +/- 4.0% and 20.8% +/- 3.41% respectively, both significantly lower than those of the none WT1-CTLs in Group B (88.9% +/- 3.4% and 91.8% +/- 5.7% respectively, both P < 0.01). There was no significant difference in the relative colony formation rate upon HLA-A * 0201(+)/WT1(-) leukemic bone marrow CD34+ progenitor cells and the HLA-A * 0201(-)/WT1(-) or HLA-A * 0201(+)/WT1(-) normal peripheral blood CD34+ progenitor cells between the CTLs of Groups A and B. CONCLUSIONS: The CTLs specific for WT1 and restricted by HLA-A * 0201 can exert specific lysis upon leukemic CD34+ progenitor cells highly expressing WT1 gene, and can inhibit the CFU-GM colony formation of such leukemic CD34+ progenitor cells. The product of expression of WT1 gene may be used as a target in the leukemic CD34+ cells.


Subject(s)
Bone Marrow Cells/immunology , Leukemia/therapy , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/genetics , Adult , Antigens, CD34 , Case-Control Studies , Cell Line, Tumor , Female , Flow Cytometry , Genes, Wilms Tumor , Granulocyte-Macrophage Progenitor Cells/immunology , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Male , Middle Aged , Peptides/genetics
20.
Zhonghua Xue Ye Xue Za Zhi ; 18(8): 403-5, 1997 Aug.
Article in Chinese | MEDLINE | ID: mdl-15625844

ABSTRACT

OBJECTIVE: To evaluate the engrafting characteristics of human mixed cord bloods. METHODS: Single or double human cord bloods were transplanted into sublethally irradiated severe combined immunodeficient (SCID) mice. Engrafting rate, hematopoiesis reconstitution and immunologic functions between the two groups were compared. RESULTS: The survival rates of both transplanted groups were equivalent (P > 0.05) while the total volume of transfused cord bloods had no statistical differences. But the engrafting rate of mixed cord blood transplant (MCBT) group was lower than that of single cord blood transplant (SCBT) group. The lymphocyte transformation rate of MCBT group was also lower than that of SCBT group. In MCBT group, only one of the two mixed cord bloods was engrafted in SCID mice as shown by HLA-DPB gene detection. CONCLUSION: Two HLA-mismatched human cord bloods transplanted in SCID mice could reconstitute hematopoietic and immunologic functions, but only one cord blood with the higher proliferation potential was engrafted.


Subject(s)
Fetal Blood/transplantation , Severe Combined Immunodeficiency/blood , Severe Combined Immunodeficiency/surgery , Animals , Female , Granulocyte-Macrophage Progenitor Cells/immunology , Hematopoiesis , Humans , Infant, Newborn , Male , Mice , Severe Combined Immunodeficiency/immunology , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...