Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 45
1.
Front Immunol ; 12: 766620, 2021.
Article En | MEDLINE | ID: mdl-34966386

Neutrophils are key pathogen exterminators of the innate immune system endowed with oxidative and non-oxidative defense mechanisms. More recently, a more complex role for neutrophils as decision shaping cells that instruct other leukocytes to fine-tune innate and adaptive immune responses has come into view. Under homeostatic conditions, neutrophils are short-lived cells that are continuously released from the bone marrow. Their development starts with undifferentiated hematopoietic stem cells that pass through different immature subtypes to eventually become fully equipped, mature neutrophils capable of launching fast and robust immune responses. During severe (systemic) inflammation, there is an increased need for neutrophils. The hematopoietic system rapidly adapts to this increased demand by switching from steady-state blood cell production to emergency granulopoiesis. During emergency granulopoiesis, the de novo production of neutrophils by the bone marrow and at extramedullary sites is augmented, while additional mature neutrophils are rapidly released from the marginated pools. Although neutrophils are indispensable for host protection against microorganisms, excessive activation causes tissue damage in neutrophil-rich diseases. Therefore, tight regulation of neutrophil homeostasis is imperative. In this review, we discuss the kinetics of neutrophil ontogenesis in homeostatic conditions and during emergency myelopoiesis and provide an overview of the different molecular players involved in this regulation. We substantiate this review with the example of an autoinflammatory disease, i.e. systemic juvenile idiopathic arthritis.


Arthritis, Juvenile/immunology , Granulocytes/immunology , Homeostasis/immunology , Leukopoiesis/immunology , Neutrophils/immunology , Bone Marrow/immunology , Bone Marrow/metabolism , Cytokines/immunology , Cytokines/metabolism , Granulocytes/cytology , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Inflammation/immunology , Inflammation/metabolism , Neutrophils/cytology
2.
J Immunol ; 205(10): 2786-2794, 2020 11 15.
Article En | MEDLINE | ID: mdl-32998984

BP180 (also termed type XVII collagen) is a hemidesmosomal protein and plays a critical role in cell-cell matrix adhesion in the skin; however, its other biological functions are largely unclear. In this study, we generated a BP180 functional-deficient mouse strain by deleting its extracellular domain of humanized NC16A (termed ΔNC16A mice). We found that BP180 is expressed by bone marrow mesenchymal stem cells (BM-MSC), and its functional deficiency leads to myeloid hyperplasia. Altered granulopoiesis in ΔNC16A mice is through bone marrow stromal cells evidenced by bone marrow transplantation. Furthermore, the level of G-CSF in bone marrow and circulation were significantly increased in ΔNC16A mice as compared with wild-type mice. The increased G-CSF was accompanied by an increased activation of the NF-κB signaling pathway in bone marrow and BM-MSC of ΔNC16A mice. Blockade of G-CSF restored normal granulopoiesis in ΔNC16A mice. Inhibition of NF-κB signaling pathway significantly reduces the release of G-CSF from ΔNC16A BM-MSC in vitro and the level of serum G-CSF in ΔNC16A mice. To our knowledge, these findings provide the first direct evidence that BP180 plays an important role in granulopoiesis through regulating NF-κB signaling pathway in BM-MSC.


Autoantigens/metabolism , Bone Marrow/pathology , Leukopoiesis/immunology , Mesenchymal Stem Cells/metabolism , Neutrophils/physiology , Non-Fibrillar Collagens/metabolism , Animals , Autoantigens/genetics , Bone Marrow/drug effects , Bone Marrow/metabolism , Cell Differentiation/immunology , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Hyperplasia/genetics , Hyperplasia/immunology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Non-Fibrillar Collagens/genetics , Protein Domains/genetics , Signal Transduction/drug effects , Signal Transduction/immunology , Collagen Type XVII
3.
Front Immunol ; 10: 1912, 2019.
Article En | MEDLINE | ID: mdl-31474989

Neutrophils are implicated in almost every stage of oncogenesis and paradoxically display anti- and pro-tumor properties. Accumulating evidence indicates that neutrophils display diversity in their phenotype resulting from functional plasticity and/or changes to granulopoiesis. In cancer, neutrophils at a range of maturation stages can be identified in the blood and tissues (i.e., outside of their developmental niche). The functional capacity of neutrophils at different states of maturation is poorly understood resulting from challenges in their isolation, identification, and investigation. Thus, the impact of neutrophil maturity on cancer progression and therapy remains enigmatic. In this review, we discuss the identification, prevalence, and function of immature and mature neutrophils in cancer and the potential impact of this on tumor progression and cancer therapy.


Cell Differentiation/immunology , Hematopoietic Stem Cells/immunology , Leukopoiesis/immunology , Neoplasms/immunology , Neutrophils/immunology , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/immunology , CCAAT-Binding Factor/metabolism , Cell Differentiation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukopoiesis/genetics , Neoplasms/genetics , Neoplasms/therapy , Neutrophils/cytology , Neutrophils/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/immunology , Trans-Activators/metabolism
4.
Bull Exp Biol Med ; 166(3): 344-347, 2019 Jan.
Article En | MEDLINE | ID: mdl-30627910

The role of signaling molecules in synthesis of humoral regulators of granulocytopoiesis by the hematopoietic microenvironmental cells during stress was analyzed using specific inhibitors. The major role in stimulation of the synthesis of granulocytic CSF during stressful stimulation is played by PI3K/Akt signaling cascade. Nuclear transcription factor NF-κB plays an auxiliary role in the regulation of functional activity of the bone marrow mononuclears. However, this factor affects the synthesis of granulocytic CSF by CD4+ cells of the bone marrow in response to stressful stimulation. Different degree and specific character of involvement of the signaling proteins in the regulation of the production of humoral factors determining colony-stimulating activity are explained by changes in functional state of monocyte-derived macrophages in different periods of stress response.


Granulocyte Colony-Stimulating Factor/genetics , Granulocytes/immunology , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/immunology , Stress, Psychological/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Chromones/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation , Gold Sodium Thiomalate/pharmacology , Granulocyte Colony-Stimulating Factor/immunology , Granulocytes/drug effects , Granulocytes/pathology , Imidazoles/pharmacology , Immobilization/methods , Leukopoiesis/drug effects , Leukopoiesis/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Phosphatidylinositol 3-Kinases/immunology , Phosphoinositide-3 Kinase Inhibitors , Pyridines/pharmacology , Stress, Psychological/immunology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/immunology
5.
Front Immunol ; 9: 349, 2018.
Article En | MEDLINE | ID: mdl-29535725

Activation and reprogramming of hematopoietic stem/progenitor cells play a critical role in the granulopoietic response to bacterial infection. Our current study determined the significance of Sonic hedgehog (SHH) signaling in the regulation of hematopoietic precursor cell activity during the host defense response to systemic bacterial infection. Bacteremia was induced in male Balb/c mice via intravenous injection (i.v.) of Escherichia coli (5 × 107 CFUs/mouse). Control mice received i.v. saline. SHH protein level in bone marrow cell (BMC) lysates was markedly increased at both 24 and 48 h of bacteremia. By contrast, the amount of soluble SHH ligand in marrow elutes was significantly reduced. These contrasting alterations suggested that SHH ligand release from BMCs was reduced and/or binding of soluble SHH ligand to BMCs was enhanced. At both 12 and 24 h of bacteremia, SHH mRNA expression by BMCs was significantly upregulated. This upregulation of SHH mRNA expression was followed by a marked increase in SHH protein expression in BMCs. Activation of the ERK1/2-SP1 pathway was involved in mediating the upregulation of SHH gene expression. The major cell type showing the enhancement of SHH expression in the bone marrow was lineage positive cells. Gli1 positioned downstream of the SHH receptor activation serves as a key component of the hedgehog (HH) pathway. Primitive hematopoietic precursor cells exhibited the highest level of baseline Gli1 expression, suggesting that they were active cells responding to SHH ligand stimulation. Along with the increased expression of SHH in the bone marrow, expression of Gli1 by marrow cells was significantly upregulated at both mRNA and protein levels following bacteremia. This enhancement of Gli1 expression was correlated with activation of hematopoietic stem/progenitor cell proliferation. Mice with Gli1 gene deletion showed attenuation in activation of marrow hematopoietic stem/progenitor cell proliferation and inhibition of increase in blood granulocytes following bacteremia. Our results indicate that SHH signaling is critically important in the regulation of hematopoietic stem/progenitor cell activation and reprogramming during the granulopoietic response to serious bacterial infection.


Escherichia coli Infections/immunology , Escherichia coli/immunology , Hedgehog Proteins/immunology , Hematopoietic Stem Cells/immunology , Leukopoiesis/immunology , Signal Transduction/immunology , Animals , Bacteremia/immunology , Bacteremia/pathology , Escherichia coli Infections/pathology , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/pathology , Male , Mice , Mice, Inbred BALB C , Zinc Finger Protein GLI1/immunology
6.
Vet Immunol Immunopathol ; 183: 40-44, 2017 Jan.
Article En | MEDLINE | ID: mdl-28063475

Flow cytometry was used to identify mAbs that recognize conserved epitopes on hamster leukocyte differentiation molecules (hLDM) and also to characterize mAbs developed against hLDM. Initial screening of mAbs developed against LDMs in other species yielded mAbs specific for the major histocompatibility (MHC) II molecule, CD4 and CD18. Screening of sets of mAbs developed against hLDM yielded 22 new mAbs, including additional mAbs to MHC II molecules and mAbs that recognize LDMs expressed on all leukocytes, granulocytes, all lymphocytes, all T cells, a subset of T cells, or on all B cells. Based on comparison of the pattern of expression of LDMs expressed on all hamster leukocytes with the patterns of expression of known LDMs in other species, as detected by flow cytometry (FC), four mAbs are predicted to recognize CD11a, CD44, and CD45. Cross comparison of mAbs specific for a subset of hamster T cells with a cross reactive mAb known to recognize CD4 in mice and one recognising CD8 revealed they recognize CD4. The characterization of these mAbs expands opportunities to use hamsters as an additional model species to investigate the mechanisms of immunopathogenesis of infectious diseases.


Antibodies, Monoclonal/immunology , Antigens, Differentiation/immunology , Cricetinae/immunology , Leukocytes/immunology , Animals , Antibody Specificity , Female , Flow Cytometry/veterinary , Leukocytes/cytology , Leukopoiesis/immunology , Male
7.
BMC Cancer ; 15: 122, 2015 Mar 13.
Article En | MEDLINE | ID: mdl-25886605

BACKGROUND: We performed a multicenter, double-blind, placebo-controlled, phase II clinical trial of human dsDNA-based preparation Panagen in a tablet form. In total, 80 female patients with stage II-IV breast cancer were recruited. METHODS: Patients received three consecutive FAC (5-fluorouracil, doxorubicin and cyclophosphamide) or AC (doxorubicin and cyclophosphamide) adjuvant chemotherapies (3 weeks per course) and 6 tablets of 5 mg Panagen or placebo daily (one tablet every 2-3 hours, 30 mg/day) for 18 days during each chemotherapy course. Statistical analysis was performed using Statistica 6.0 software, and non-parametric analyses, namely Wilcoxon-Mann-Whitney and paired Wilcoxon tests. To describe the results, the following parameters were used: number of observations (n), median, interquartile range, and minimum-maximum range. RESULTS: Panagen displayed pronounced leukostimulatory and leukoprotective effects when combined with chemotherapy. In an ancillary protocol, anticancer effects of a tablet form of Panagen were analyzed. We show that Panagen helps maintain the pre-therapeutic activity level of innate antitumor immunity and induces formation of a peripheral pool of cytotoxic CD8+ perforin + T-cells. Our 3-year follow-up analysis demonstrates that 24% of patients who received Panagen relapsed or died after the therapy, as compared to 45% in the placebo cohort. CONCLUSIONS: The data collected in this trial set Panagen as a multi-faceted "all-in-one" medicine that is capable of simultaneously sustaining hematopoiesis, sparing the innate immune cells from adverse effects of three consecutive rounds of chemotherapy and boosting individual adaptive immunity. Its unique feature is that it is delivered via gastrointestinal tract and acts through the lymphoid system of intestinal mucosa. Taken together, maintenance of the initial levels of innate immunity, development of adaptive cytotoxic immune response and significantly reduced incidence of relapses 3 years after the therapy argue for the anticancer activity of Panagen. TRIAL REGISTRATION: ClinicalTrials.gov NCT02115984 from 04/07/2014.


Adaptive Immunity/drug effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , DNA/administration & dosage , Leukopoiesis/drug effects , Adaptive Immunity/immunology , Breast Neoplasms/immunology , DNA/chemistry , Double-Blind Method , Female , Follow-Up Studies , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukopoiesis/immunology
8.
PLoS One ; 9(4): e90227, 2014.
Article En | MEDLINE | ID: mdl-24691464

This work studied the effect of protein malnutrition on the hemato-immune response to the respiratory challenge with Streptococcus pneumoniae and evaluated whether the dietary recovery with a probiotic strain has a beneficial effect in that response. Three important conclusions can be inferred from the results presented in this work: a) protein-malnutrition significantly impairs the emergency myelopoiesis induced by the generation of the innate immune response against pneumococcal infection; b) repletion of malnourished mice with treatments including nasally or orally administered Lactobacillus rhamnosus CRL1505 are able to significantly accelerate the recovery of granulopoiesis and improve innate immunity and; c) the immunological mechanisms involved in the protective effect of immunobiotics vary according to the route of administration. The study demonstrated that dietary recovery of malnourished mice with oral or nasal administration of L. rhamnosus CRL1505 improves emergency granulopoiesis and that CXCR4/CXCR12 signaling would be involved in this effect. Then, the results summarized here are a starting point for future research and open up broad prospects for future applications of probiotics in the recovery of immunocompromised malnourished hosts.


Dietary Supplements , Immunity, Innate/immunology , Lacticaseibacillus rhamnosus/immunology , Leukopoiesis/immunology , Lung/immunology , Protein-Energy Malnutrition/immunology , Protein-Energy Malnutrition/microbiology , Animals , Bone Marrow/pathology , Bronchoalveolar Lavage Fluid , Cytokines/blood , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Myeloid Cells/pathology , Pneumococcal Infections/blood , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Protein-Energy Malnutrition/blood
9.
Blood ; 121(3): 519-29, 2013 Jan 17.
Article En | MEDLINE | ID: mdl-23212517

Neutrophils are critically involved in host defense and tissue damage. Intrinsic molecular mechanisms controlling neutrophil differentiation and activities are poorly defined. Herein we found that p53-induced phosphatase 1(Wip1) is preferentially expressed in neutrophils among immune cells. The Wip1 expression is gradually up-regulated during the differentiation of myeloid precursors into mature neutrophils. Wip1-deficient mice and chimera mice with Wip1(-/-) hematopoietic cells had an expanded pool of neutrophils with hypermature phenotypes in the periphery. The in vivo and in vitro studies showed that Wip1 deficiency mainly impaired the developing process of myeloid progenitors to neutrophils in an intrinsic manner. Mechanism studies showed that the enhanced development and maturation of neutrophils caused by Wip1 deficiency were mediated by p38 MAPK-STAT1 but not p53-dependent pathways. Thus, our findings identify a previously unrecognized p53-independent function of Wip1 as a cell type-specific negative regulator of neutrophil generation and homeostasis through limiting the p38 MAPK-STAT1 pathway.


MAP Kinase Signaling System/immunology , Neutrophils/cytology , Neutrophils/enzymology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Animals , Cell Differentiation/immunology , Female , Homeostasis/immunology , Leukocyte Disorders/congenital , Leukocyte Disorders/genetics , Leukocyte Disorders/immunology , Leukopoiesis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/enzymology , Myeloid Progenitor Cells/immunology , Neutrophils/immunology , Protein Phosphatase 2C , Respiratory Burst/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Semin Hematol ; 49(2): 138-48, 2012 Apr.
Article En | MEDLINE | ID: mdl-22449624

Hypereosinophilia, defined as peripheral blood eosinophil counts > 1,500/µL, may complicate the course of various lymphoproliferative disorders. Among these, Hodgkin lymphoma (HL) and certain peripheral T-cell lymphomas (PTCLs) derived from CD4 cells, including Sezary syndrome (SS), adult T-cell leukemia/lymphoma (ATLL), and angioimmunoblastic T-cell lymphoma (AITL), are most commonly associated with increased reactive eosinophilopoiesis. Rarely, marked hypereosinophilia (HE) may occur in the setting of acute B-cell lymphoblastic leukemia, with a substantial impact on disease course. The mechanisms leading to blood and tissue eosinophilia in the setting of lymphoproliferative disorders, as well as the clinical complications and prognostic implications of hypereosinophilia, are discussed in this review.


Hypereosinophilic Syndrome/immunology , Lymphoproliferative Disorders/immunology , Animals , Humans , Hypereosinophilic Syndrome/pathology , Leukopoiesis/immunology , Lymphoproliferative Disorders/pathology
11.
Blood ; 118(23): 6172-82, 2011 Dec 01.
Article En | MEDLINE | ID: mdl-21972291

The mechanisms by which innate immune signals regulate alloimmune responses remain poorly understood. In the present study, we show by intravital 2-photon microscopy direct interactions between graft-infiltrating neutrophils and donor CD11c(+) dendritic cells (DCs) within orthotopic lung allografts immediately after reperfusion. Neutrophils isolated from the airways of lung transplantation recipients stimulate donor DCs in a contact-dependent fashion to augment their production of IL-12 and expand alloantigen-specific IFN-γ(+) T cells. DC IL-12 expression is largely regulated by degranulation and induced by TNF-α associated with the neutrophil plasma membrane. Extended cold ischemic graft storage enhances G-CSF-mediated granulopoiesis and neutrophil graft infiltration, resulting in exacerbation of ischemia-reperfusion injury after lung transplantation. Ischemia reperfusion injury prevents immunosuppression-mediated acceptance of mouse lung allografts unless G-CSF-mediated granulopoiesis is inhibited. Our findings identify granulopoiesis-mediated augmentation of alloimmunity as a novel link between innate and adaptive immune responses after organ transplantation.


Dendritic Cells/cytology , Graft Rejection/immunology , Leukopoiesis/immunology , Lung Transplantation/immunology , Neutrophils/cytology , Acute Disease , Animals , Cell Degranulation/immunology , Cell Membrane/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Interleukin-12/metabolism , Leukopoiesis/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/metabolism , Reperfusion Injury/immunology , Signal Transduction/immunology , Transplantation Immunology/immunology , Transplantation, Homologous , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
PLoS One ; 6(5): e19957, 2011.
Article En | MEDLINE | ID: mdl-21655273

Normally, neutrophil pools are maintained by homeostatic mechanisms that require the transcription factor C/EBPα. Inflammation, however, induces neutrophilia through a distinct pathway of "emergency" granulopoiesis that is dependent on C/EBPß. Here, we show in mice that alum triggers emergency granulopoiesis through the IL-1RI-dependent induction of G-CSF. G-CSF/G-CSF-R neutralization impairs proliferative responses of hematopoietic stem and progenitor cells (HSPC) to alum, but also abrogates the acute mobilization of BM neutrophils, raising the possibility that HSPC responses to inflammation are an indirect result of the exhaustion of BM neutrophil stores. The induction of neutropenia, via depletion with Gr-1 mAb or myeloid-specific ablation of Mcl-1, elicits G-CSF via an IL-1RI-independent pathway, stimulating granulopoietic responses indistinguishable from those induced by adjuvant. Notably, C/EBPß, thought to be necessary for enhanced generative capacity of BM, is dispensable for increased proliferation of HSPC to alum or neutropenia, but plays a role in terminal neutrophil differentiation during granulopoietic recovery. We conclude that alum elicits a transient increase in G-CSF production via IL-1RI for the mobilization of BM neutrophils, but density-dependent feedback sustains G-CSF for accelerated granulopoiesis.


Granulocyte Colony-Stimulating Factor/blood , Granulocytes/immunology , Inflammation/immunology , Leukopoiesis/immunology , Alum Compounds/toxicity , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Granulocytes/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Inflammation/blood , Inflammation/chemically induced , Leukopoiesis/drug effects , Mice , Mice, Inbred C57BL , Neutropenia/blood , Neutropenia/chemically induced , Neutropenia/immunology
13.
J Immunol ; 186(5): 2826-34, 2011 Mar 01.
Article En | MEDLINE | ID: mdl-21270394

Tristetraprolin (TTP, Zfp36, Nup475, Tis11) dramatically reduces the stability of target mRNAs by binding to AU-rich elements in their 3' untranslated regions. Through this mechanism, TTP functions as a rheostatic, temporal regulator of gene expression. TTP knockout (KO) mice exhibit completely penetrant granulocytic hyperplasia. We have shown that the hematopoietic stem-progenitor cell compartment in TTP KO mice is also altered. Although no change was detected in long-term hematopoietic stem cell (HSC) frequency or function, as assayed by immunophenotypic markers or limiting dilution transplants, we observed increases in the frequencies and numbers of short-term HSCs, multipotent progenitors, and granulocyte-monocyte progenitors. This pattern is consistent with "reactive granulopoiesis," in which committed myeloid progenitors and more primitive progenitors cycle more actively to increase production of mature granulocytes in response to infection or adjuvant. We created reverse chimeras by transplanting wild-type bone marrow into TTP KO mice and found the "reactive granulopoiesis" phenocopied, indicating a non-hematopoietic stem-progenitor cell-autonomous mechanism. Correspondingly, we found elevated levels of the granulopoietic TTP targets IL-1ß, TNF-α, and IL-6 in the plasma of TTP KO mice. Consistent with the non-cell-autonomous nature of the phenotype, we found elevated levels of IL-1ß, TNF-α, and IL-6 transcripts in the livers of TTP KO mice and no detectable difference in the bone marrows. These findings demonstrate the importance of TTP in inflammatory homeostasis and highlight the ability of the hematopoietic system to respond to stress without significant numbers of quiescent HSCs entering the cell cycle.


Granulocytes/immunology , Hematopoietic Stem Cells/immunology , Leukopoiesis/immunology , Resting Phase, Cell Cycle/immunology , Tristetraprolin/deficiency , Tristetraprolin/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Cycle/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/metabolism , Female , Granulocytes/metabolism , Granulocytes/pathology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , Homeostasis/immunology , Immunophenotyping , Leukopoiesis/genetics , Liver/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Resting Phase, Cell Cycle/genetics , Tristetraprolin/physiology , Up-Regulation/immunology
14.
Immunobiology ; 216(1-2): 213-7, 2011.
Article En | MEDLINE | ID: mdl-20965099

As the inbred mouse strain SJL/J displays increased resistance to several pathogens and as its immune system shows multiple specificities, it is tempting to infer a causal link between these observations. The first question that comes to mind is whether adaptive immunity plays a role, and a way to answer this question is to see if the resistance phenotype persists when adaptive immunity is depressed. Although it has long been known that irradiation causes repression of leukopoiesis in mice, the technical data available in the literature are of no help in the case of strain SJL/J, because it displays exceptional radioresistance. Here we show that exposure of SJL/J to ∼9Gy, an intensity corresponding to the lethal dose 50 for the species Mus musculus, leads to serious but reversible alteration of leukopoiesis. This conclusion stems from an examination of the effects, 1-11 days post-exposure, of whole-body gamma-ray irradiation on leukocyte populations in the thymus and peripheral blood of young adult females. Immunodepression was most severe 4 days post-exposure. As in other strains, leukocyte populations displayed differential radiosensitivity, B (CD19(+)) cells being most sensitive, T (CD4(+)/CD8(+)) cells moderately sensitive, and natural killer (NK1.1(+)) cells most resistant. Surprisingly, however, the helper/inducer T lymphocytes proved more resistant than the cytotoxic/suppressor T lymphocytes, contrarily to what is observed in other strains. The procedure described will make it possible to refute or establish reliably the existence of causal links between SJL-specific phenotypic traits and immune aberrations and to elucidate further the respective roles of innate and acquired immunity in determining the resistance of this strain to an array of viral diseases.


B-Lymphocytes/radiation effects , Immunity, Active , Immunosuppression Therapy , T-Lymphocytes/radiation effects , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Count , Female , Gamma Rays/adverse effects , Immunity, Active/radiation effects , Lethal Dose 50 , Leukopoiesis/immunology , Leukopoiesis/radiation effects , Mice , Mice, Inbred Strains , Radiation Tolerance/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Whole-Body Irradiation
15.
Blood ; 116(19): 3955-63, 2010 Nov 11.
Article En | MEDLINE | ID: mdl-20682855

The development of the mononuclear phagocyte system requires macrophage colony-stimulating factor (CSF-1) signaling through the CSF-1 receptor (CSF1R, CD115). We examined the effect of an antibody against CSF1R on macrophage homeostasis and function using the MacGreen transgenic mouse (csf1r-enhanced green fluorescent protein) as a reporter. The administration of a novel CSF1R blocking antibody selectively reduced the CD115(+)Gr-1(neg) monocyte precursor of resident tissue macrophages. CD115(+)Gr-1(+) inflammatory monocytes were correspondingly increased, supporting the view that monocytes are a developmental series. Within tissue, the antibody almost completely depleted resident macrophage populations in the peritoneum, gastrointestinal tract, liver, kidney, and skin, but not in the lung or female reproductive organs. CSF1R blockade reduced the numbers of tumor-associated macrophages in syngeneic tumor models, suggesting that these cells are resident type macrophages. Conversely, it had no effect on inflammatory monocyte recruitment in models, including lipopolysaccharide-induced lung inflammation, wound healing, peritonitis, and severe acute graft-versus-host disease. Depletion of resident tissue macrophages from bone marrow transplantation recipients actually resulted in accelerated pathology and exaggerated donor T-cell activation. The data indicate that CSF1R signaling is required only for the maturation and replacement of resident-type monocytes and tissue macrophages, and is not required for monocyte production or inflammatory function.


Antibodies, Monoclonal/pharmacology , Inflammation/immunology , Macrophages/immunology , Monocytes/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Cell Line, Tumor , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Inflammation/pathology , Inflammation/therapy , Leukopoiesis/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/classification , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Rats
16.
Kidney Int ; 78(1): 79-88, 2010 Jul.
Article En | MEDLINE | ID: mdl-20375992

Mycophenolic acid is a commonly used immunosuppressant after organ transplantation and in autoimmune diseases; however, myelosuppression is a major complication despite its largely favorable side-effect profile. Mycophenolic acid targets inosine monophosphate dehydrogenase, which is essential for T-cell proliferation. The T-cell cytokine interleukin-17 (IL-17 or IL-17A) and its receptor maintain normal neutrophilic granulocyte numbers in mice by induction of granulocyte-colony-stimulating factor. To test whether mycophenolic acid induces neutropenia by inhibiting IL-17-producing T cells, we treated C57Bl/6 mice with mycophenolate-mofetil (the orally available pro-drug) and found a dose-dependent decrease in blood neutrophils. This myelosuppressive effect was completely abolished in mice that lack the IL-17 receptor. Mycophenolic acid delayed myeloid recovery after bone marrow transplantation and decreased the percentage of IL-17-producing T cells in the spleen and thymus, and inhibited IL-17 production in human and mouse T cells in vitro. Injection of IL-17 during mycophenolic acid treatment overcame the suppression of the circulating neutrophil levels. Our study shows that mycophenolic acid suppresses neutrophil production by inhibiting IL-17 expression, suggesting that measurement of this interleukin might be useful in estimating the risk of neutropenia in clinical settings.


Interleukin-17/metabolism , Mycophenolic Acid/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Colony-Stimulating Factors/immunology , Colony-Stimulating Factors/pharmacology , Cytokines/immunology , Cytokines/pharmacology , Female , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/pharmacology , Interleukin-17/immunology , Interleukin-17/pharmacology , Leukopoiesis/drug effects , Leukopoiesis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/immunology , Neutropenia/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Interleukin-17/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Blood ; 113(17): 3990-8, 2009 Apr 23.
Article En | MEDLINE | ID: mdl-19088377

The Rac1 and Rac2 GTPases play important roles in many processes including cytoskeletal reorganization, proliferation, and survival, and are required for B-cell development. Previous studies had shown that deficiency in Rac2 did not affect T-cell development, whereas the function of Rac1 in this process has not been investigated. We now show that simultaneous absence of both GTPases resulted in a very strong developmental block at the pre-TCR checkpoint and in defective positive selection. Unexpectedly, deficiency of Rac1 and Rac2 also resulted in the aberrant survival of thymocytes lacking expression of TCR beta, showing hallmarks of hyperactive Notch signaling. Furthermore, we found a similar novel phenotype in the absence of Vav1, Vav2, and Vav3, which function as guanine nucleotide exchange factors for Rac1 and Rac2. These results show that a pathway containing Vav and Rac proteins may negatively regulate Notch signaling during early thymic development.


Leukopoiesis/immunology , Neuropeptides/metabolism , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , rac GTP-Binding Proteins/metabolism , Animals , Cell Proliferation , Humans , Interleukin-7/metabolism , Mice , Mice, Knockout , Neuropeptides/deficiency , Neuropeptides/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Notch/metabolism , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymus Gland/enzymology , Thymus Gland/immunology , Time Factors , Tissue Culture Techniques , rac GTP-Binding Proteins/deficiency , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein , RAC2 GTP-Binding Protein
18.
Inflamm Bowel Dis ; 14(11): 1491-503, 2008 Nov.
Article En | MEDLINE | ID: mdl-18623170

BACKGROUND: Chronic inflammatory diseases are characterized by massive infiltration of innate and acquired immune cells in inflammatory sites. However, it remains unclear how these cells cooperate in the development of disease. Although bone marrow (BM) is a primary site for hematopoiesis of immune cells except T cells, BM recruits memory T cells from the periphery. We have recently demonstrated that colitogenic CD4(+) memory T cells reside in BM of colitic CD4(+)CD45RB(high) T-cell-transferred SCID mice. Based on this background we here investigate whether granulocytes promote or suppress the expansion of colitogenic CD4(+) T cells. METHODS: First, we show that Gr-1(high)CD11b(+) granulocytes were significantly increased in colitic BM along with a significant increase of peripheral granulocytes. Consistently, the colony-forming unit (CFU) assay revealed that granulocyte colony formation was dominantly induced by supernatants from anti-CD3-stimulated colitic BM CD4(+) T cells. RESULTS: Administration of granulocyte-depleting anti-Gr-1 mAb to colitic mice did not ameliorate the colitis, but exacerbated the wasting disease with an increased expansion of systemic, but not lamina propria, CD4(+) T cells with activated phenotype. CONCLUSIONS: These results suggest that the increased granulopoiesis by colitogenic BM CD4(+) T cells represent a negative feedback mechanism to control systemic inflammation.


CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Feedback, Physiological/immunology , Granulocytes/immunology , Leukopoiesis/immunology , Animals , CD4-Positive T-Lymphocytes/physiology , Cells, Cultured , Chronic Disease , Colitis/physiopathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Feedback, Physiological/physiology , Flow Cytometry , Granulocytes/physiology , Immunohistochemistry , Leukopoiesis/physiology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, SCID , Probability , Random Allocation , Sensitivity and Specificity , Splenectomy , Statistics, Nonparametric
19.
Allergy ; 62(12): 1410-8, 2007 Dec.
Article En | MEDLINE | ID: mdl-17983375

BACKGROUND: The mechanisms of the distant stimulation of the bone marrow (BM) after airway allergen exposure remain largely obscure. T cells have been implicated in allergic airway inflammation but their role in allergen-induced BM eosinophilopoiesis is poorly understood. The aim of this study was to determine the role of CD4(+) and CD8(+) T cells in allergen-induced BM eosinophilopoiesis. METHODS: Ovalbumin (OVA)-sensitized wild type (WT), CD4 knockout (CD4-/-) and CD8 knockout (CD8-/-) mice were exposed intranasally to OVA or saline. Bromo-deoxyuridine (BrdU) was used to label newly produced cells. Bone marrow, blood and bronchoalveolar lavage (BAL) were sampled 24 h after the final exposure. Immunostaining for newly produced eosinophils (i.e. BrdU(+)/MBP(+)) and BM eosinophil progenitor [CD34(+)/CD45(+)/interleukin-5 (IL-5)Ralpha(+)] cells was performed. RESULTS: The number of newly produced BM eosinophils (BrdU(+)/MBP(+) cells) was significantly reduced in allergen exposed CD4-/- or CD8-/- mice compared with allergen exposed WT mice, which was followed by a subsequent decrease in newly produced blood and airway eosinophils. Furthermore, BM eosinophil progenitors were significantly reduced in allergen exposed CD4-/- and CD8-/- mice compared with WT mice. Finally, serum IL-5 and Bronchoalveolar lavage fluid eotaxin-2 levels were abolished in allergen exposed CD4-/- mice to levels seen in saline exposed WT mice. CONCLUSIONS: These data suggests that both CD4(+) and CD8(+) T cells have a regulatory role in allergen-induced BM eosinophilopoiesis, whereas CD4(+) T cells are obligatory for allergen-induced airway eosinophilia. The subsequent traffic of eosinophils to the airways is likely to be at least partly regulated by a CD4(+) T-cell-dependent local airway eotaxin-2 production.


Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Eosinophils/immunology , Leukopoiesis/immunology , Ovalbumin/immunology , Allergens/administration & dosage , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/immunology , Bronchoalveolar Lavage Fluid/immunology , CD4 Antigens/genetics , CD8 Antigens/genetics , Chemokine CCL24/metabolism , Eosinophils/cytology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage
20.
J Immunol ; 179(9): 5793-802, 2007 Nov 01.
Article En | MEDLINE | ID: mdl-17947652

A newly recognized link between the complement system and adaptive immunity is that decay accelerating factor (DAF), a cell surface C3/C5 convertase regulator, exerts control over T cell responses. Extending these results, we show that cultures of Marilyn TCR-transgenic T cells stimulated with DAF-deficient (Daf1(-/-)) APCs produce significantly more IL-12, C5a, and IFN-gamma compared with cultures containing wild-type APCs. DAF-regulated IL-12 production and subsequent T cell differentiation into IFN-gamma-producing effectors was prevented by the deficiency of either C3 or C5a receptor (C5aR) in the APC, demonstrating a link between DAF, local complement activation, IL-12, and T cell-produced IFN-gamma. Bone marrow chimera experiments verified that bone marrow cell-expressed C5aR is required for optimal differentiation into IFN-gamma-producing effector T cells. Overall, our results indicate that APC-expressed DAF regulates local production/activation of C5a following cognate T cell/APC interactions. Through binding to its receptor on APCs the C5a up-regulates IL-12 production, this in turn, contributes to directing T cell differentiation toward an IFN-gamma-producing phenotype. The findings have implications for design of therapies aimed at altering pathologic T cell immunity.


CD55 Antigens/metabolism , Complement C5a/metabolism , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Leukopoiesis/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Bone Marrow/metabolism , CD55 Antigens/genetics , Complement C5a/genetics , Interleukin-12/deficiency , Interleukin-12/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Receptors, Interleukin-12/metabolism
...