Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 618
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cytokine ; 95: 35-42, 2017 07.
Article in English | MEDLINE | ID: mdl-28235674

ABSTRACT

In the midgestation mouse embryo, hematopoietic cell clusters containing hematopoietic stem/progenitor cells arise in the aorta-gonad-mesonephros (AGM) region. We have previously reported that forced expression of the Sox17 transcription factor in CD45lowc-Kithigh AGM cells, which are the hematopoietic cellular component of the cell clusters, and subsequent coculture with OP9 stromal cells in the presence of three cytokines, stem cell factor (SCF), interleukin-3 (IL-3), and thrombopoietin (TPO), led to the formation and the maintenance of cell clusters with cells at an undifferentiated state in vitro. In this study, we investigated the role of each cytokine in the formation of hematopoietic cell clusters. We cultured Sox17-transduced AGM cells with each of the 7 possible combinations of the three cytokines. The size and the number of Sox17-transduced cell clusters in the presence of TPO, either alone or in combination, were comparable to that observed with the complete set of the three cytokines. Expression of TPO receptor, c-Mpl was almost ubiquitously expressed and maintained in Sox17-transduced hematopoietic cell clusters. In addition, the expression level of c-Mpl was highest in the CD45lowc-Kithigh cells among the Sox17-transduced cell clusters. Moreover, c-Mpl protein was highly expressed in the intra-aortic hematopoietic cell clusters in comparison with endothelial cells of dorsal aorta. Finally, stimulation of the endothelial cells prepared from the AGM region by TPO induced the production of hematopoietic cells. These results suggest that TPO contributes to the formation and the maintenance of hematopoietic cell clusters in the AGM region.


Subject(s)
Aorta/cytology , Gonads/cytology , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mesonephros/cytology , Thrombopoietin/physiology , Animals , Aorta/embryology , Aorta/metabolism , Cells, Cultured , Gonads/embryology , Gonads/metabolism , Interleukin-3/physiology , Mesonephros/metabolism , Mice, Inbred C57BL , Mice, Inbred ICR , Receptors, Thrombopoietin/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Stem Cell Factor/physiology , Transduction, Genetic
2.
Tumour Biol ; 35(4): 3471-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24293394

ABSTRACT

The aim of this study was to identify candidate single-nucleotide polymorphisms (SNPs) that might play a role in susceptibility to neuroblastoma, elucidate their potential mechanisms, and generate SNP-to-gene-to-pathway hypotheses. A genome-wide association study (GWAS) dataset of neuroblastoma that included 442,976 SNPs from 1,627 neuroblastoma patients and 3,254 control subjects of European descent was used in this study. The identify candidate causal SNPs and pathways (ICSNPathway) analysis was applied to the GWAS dataset. ICSNPathway analysis identified 15 candidate SNPs, 10 genes, and 31 pathways, which revealed 10 hypothetical biological mechanisms. The strongest hypothetical biological mechanism was one wherein SNPrs40401 modulates the role of IL3 in several pathways and conditions, including the stem pathway, asthma (hsa05310), the dendritic cell pathway, and development (0.001 < p < 0.004; 0.001 < FDR < 0.033). The second strongest mechanism identified was that in which rs1048108 and rs16852600 alter the function of BARD1, which negatively regulates developmental process and modulates processes including cell development and programmed cell death (0.001 < p < 0.004; 0.001 < FDR < 0.033). The third mechanism identified was one wherein rs1939212 modulated CFL1, resulting in negative regulation of development, cell death, neural crest cell migration, and apoptosis (0.001 < p < 0.004; 0.001 < FDR < 0.033). By using the ICSNPathway to analyze neuroblastoma GWAS data, 15 candidate SNPs, 10 genes including IL3, BARD1, and CFL, and 31 pathways were identified that might contribute to the susceptibility of patients to neuroblastoma.


Subject(s)
Genome-Wide Association Study , Neuroblastoma/genetics , Polymorphism, Single Nucleotide , Signal Transduction , Cofilin 1/physiology , Humans , Interleukin-3/physiology , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/physiology
3.
Ann Hematol ; 93(12): 2019-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25029985

ABSTRACT

Cytokine-induced killer (CIK) cells, one of the feasible and effective methods of adoptive immunotherapy, have shown anti-leukemia activity in vivo and in vitro. But the strategy exhibits limited cytotoxic activity in clinical studies. In this study, CIK cells were transfected with an interleukin-3/Pseudomonas exotoxin gene (IL3PE38KDEL). RT-PCR and ELISA were used to verify the expression of IL3PE38KDEL in the transfected CIK cells. These cells released 1,186.7 ± 149.6 pg IL3PE38KDEL/10(4) cells over 48 h into the medium and the culture supernatant selectively killed IL3 receptor(IL3R)-positive HL60 cells, but not IL3R-negative K562 cells. Moreover, IL3PE38KDEL transfection did not influence phenotypes and cytokine production of CIK cells. Co-cultured with leukemia cells, IL3PE38KDEL transfected CIK cells showed enhanced cytotoxicity against IL3R-positive HL60 cells at all effector-to-target (E:T) ratios, but exerted a basal anti-leukemia activity against IL3R-negative K562 cells. Our findings demonstrate that IL3PE38KDEL gene transfection may be a novel strategy for improving anti-leukemia activity of CIK cells.


Subject(s)
ADP Ribose Transferases/physiology , Cytokine-Induced Killer Cells/immunology , Exotoxins/physiology , Interleukin-3/physiology , Leukemia, Myeloid, Acute/pathology , Pseudomonas aeruginosa/genetics , Transfection , Virulence Factors/physiology , ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Coculture Techniques , Cytotoxicity, Immunologic , Exotoxins/genetics , Genes, Synthetic , HL-60 Cells , Humans , Immunophenotyping , Immunotherapy , Interferon-gamma/analysis , Interleukin-3/genetics , K562 Cells , Mutation , Protein Structure, Tertiary , Tumor Necrosis Factor-alpha/analysis , Virulence Factors/genetics , Pseudomonas aeruginosa Exotoxin A
4.
J Biol Regul Homeost Agents ; 28(1): 17-21, 2014.
Article in English | MEDLINE | ID: mdl-24750787

ABSTRACT

It is well established that mast cells, which are found in the tissues in the proximity of small blood vessels and post-capillary venules, play a key role in the early phase of IgE-mediated allergic reactions. A greatly expanded understanding of the biology of IL-3 has emerged since the early 1980s. IL-3 is a specific factor that stimulates the growth of hematopoietic stem and progenitor cells of a variety of lineages and can promote the proliferation of certain classes of lymphocytes distinct from those that are dependent on IL-2. IL-3 has been identified among the most important cytokines for regulation of mast cell growth and differentiation, migration and effector function activities of many hematopoietic cells. IL-3 termed multi colony-stimulating-factor (multi-CSF) or mast cell growth factor (MCGF) is a haematopoietic growth factor which stimulates the formation of colonies for erythroid, megakaryocytic, granulocytic and monocytic lineages. It is predominantly produced by activated T cells, natural killer (NK) cells and mast cells and supports the growth-promoting effects of SCF on mast cell precursors. IL-3 causes severe hypersensivity reactions and plays a pivotal role in exacerbating the inflammatory response in vivo. Here we report the interrelationship between IL-3 and mast cells.


Subject(s)
Interleukin-3/physiology , Mast Cells/physiology , Animals , Calcium/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation/immunology
5.
J Biol Regul Homeost Agents ; 28(1): 91-103, 2014.
Article in English | MEDLINE | ID: mdl-24750795

ABSTRACT

Chronic aeroallergen inhalation elicits the expansion of IL-4-producing Th2 cells and the production of IgE antibodies. In sensitized subjects, who have established IgE and Th2 responses, re-exposure to allergen leads to rapid recruitment of basophils, which are thought to be important effectors of late phase allergic reactions. Several investigations of responses to parasites and injected antigens have identified an additional role for basophils as innate immune effectors during initial antigen encounter in immunologically naïve hosts. These cells constitutively express IL-4 and promote Th2 polarized adaptive responses to such antigens. Their early recruitment and modulation of cellular immune responses to natural inhaled allergens in the airways has been scarcely investigated. In this study, basophils were enumerated in lung tissue, blood and spleen from BALB/c mice in the first days after inhalation of an aqueous extract of the allergen, Aspergillus fumigatus (Af). Af inhalation induced rapid increases in basophil numbers in the lung, blood and spleen. This was Rag-1-, MyD88- and IL-3-independent. The basophils expressed abundant IL-4. Their depletion during Af sensitization resulted in an attenuated induction of both IL-4 producing Th lymphocytes and specific IgE and IgG1 responses to an inhaled protein antigen, ovalbumin, which was co-administered. Our results suggest that basophils are rapidly recruited to the airways of naïve mice following initial fungal allergen exposure, produce IL-4 and influence the development of the adaptive immune response.


Subject(s)
Adaptive Immunity , Allergens/immunology , Basophils/physiology , Interleukin-4/biosynthesis , Animals , Aspergillus fumigatus/immunology , Cell Movement , Interleukin-3/physiology , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/physiology , Neutrophils/physiology , Th2 Cells/immunology
6.
Exp Cell Res ; 319(17): 2728-38, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23933517

ABSTRACT

We have shown previously that mitochondrial ROS production is essential to turn growth factor (GF) removal into cell death. Activated RAF, AKT, Bcl-2 and antioxidants protected equally well against ROS accumulation and subsequent death. Here we investigated whether protection by survival signaling and antioxidants utilizes shared or distinct targets. Using serum deprivation from NIH 3T3 fibroblasts and IL-3 withdrawal from promyeloid 32D cells, we showed that pro-survival signaling by activated RAF but not AKT prevented the decline in Mcl-1 following GF abrogation. GF starvation increased levels of Bim in both model systems, which was prevented by RAF in 32D cells but not in NIH 3T3 fibroblasts. RAF and AKT suppressed activation and mitochondrial translocation of BAX. Also, antioxidant treatment efficiently prevented BAX activation and death of 32D cells but showed little effect on its mitochondrial translocation. No significant impact of antioxidant treatment on Bim or Mcl-1 expression was observed. ROS produced during GF abrogation also did not alter the activity of intracellular signaling pathways, which have been implicated previously in cell killing by pro-oxidants. Together these data suggest Bcl-2 family proteins as convergence point for RAF and ROS in life and death decisions.


Subject(s)
Antioxidants/pharmacology , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism , raf Kinases/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Culture Media, Serum-Free , Interleukin-3/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , NIH 3T3 Cells , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism
7.
Biochem Biophys Res Commun ; 440(4): 545-50, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24103757

ABSTRACT

Interleukin (IL)-3, a multilineage hematopoietic growth factor, is implicated in the regulation of osteoclastogenesis. However, the role of IL-3 in osteoclastogenesis remains controversial; whereas early studies showed that IL-3 stimulates osteoclastogenesis, recent investigations demonstrated that IL-3 inhibits osteoclast formation. The objective of this work is to further address the role of IL-3 in osteoclastogenesis. We found that IL-3 treatment of bone marrow cells generated a population of cells capable of differentiating into osteoclasts in tissue culture dishes in response to the stimulation of the monocyte/macrophage-colony stimulating factor (M-CSF) and the receptor activator of nuclear factor kappa B ligand (RANKL). The IL-3-dependent hematopoietic cells were able to further proliferate and differentiate in response to M-CSF stimulation and the resulting cells were also capable of forming osteoclasts with M-CSF and RANKL treatment. Interestingly, IL-3 inhibits M-CSF-/RANKL-induced differentiation of the IL-3-dependent hematopoietic cells into osteoclasts. The flow cytometry analysis indicates that while IL-3 treatment of bone marrow cells slightly affected the percentage of osteoclast precursors in the surviving populations, it considerably increased the percentage of osteoclast precursors in the populations after subsequent M-CSF treatment. Moreover, osteoclasts derived from IL-3-dependent hematopoietic cells were fully functional. Thus, we conclude that IL-3 plays dual roles in osteoclastogenesis by promoting the development of osteoclast progenitors but inhibiting the osteoclastogenic process. These findings provide a better understanding of the role of IL-3 in osteoclastogenesis.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Interleukin-3/physiology , Osteoclasts/cytology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Hematopoietic Stem Cells/drug effects , Interleukin-3/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL
8.
J Immunol ; 186(1): 516-26, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21106848

ABSTRACT

Human blood eosinophils exhibit a hyperactive phenotype in response to chemotactic factors after cell "priming" with IL-5 family cytokines. Earlier work has identified ERK1/2 as molecular markers for IL-5 priming, and in this article, we show that IL-3, a member of the IL-5 family, also augments fMLP-stimulated ERK1/2 phosphorylation in primary eosinophils. Besides ERK1/2, we also observed an enhancement of chemotactic factor-induced Akt phosphorylation after IL-5 priming of human blood eosinophils. Administration of a peptide antagonist that targets the Src family member Lyn before cytokine (IL-5/IL-3) priming of blood eosinophils inhibited the synergistic increase of fMLP-induced activation of Ras, ERK1/2 and Akt, as well as the release of the proinflammatory factor leukotriene C(4). In this study, we also examined a human eosinophil-like cell line HL-60 clone-15 and observed that these cells exhibited significant surface expression of IL-3Rs and GM-CSFRs, as well as ERK1/2 phosphorylation in response to the addition of IL-5 family cytokines or the chemotactic factors fMLP, CCL5, and CCL11. Consistent with the surface profile of IL-5 family receptors, HL-60 clone-15 recapitulated the enhanced fMLP-induced ERK1/2 phosphorylation observed in primary blood eosinophils after priming with IL-3/GM-CSF, and small interfering RNA-mediated knockdown of Lyn expression completely abolished the synergistic effects of IL-3 priming on fMLP-induced ERK1/2 phosphorylation. Altogether, our data demonstrate a central role for Lyn in the mechanisms of IL-5 family priming and suggest that Lyn contributes to the upregulation of the Ras-ERK1/2 and PI3K-Akt cascades, as well as the increased leukotriene C(4) release observed in response to fMLP in "primed" eosinophils.


Subject(s)
Eosinophils/immunology , Interleukin-3/physiology , Interleukin-5/physiology , Leukotriene C4/metabolism , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , ras Proteins/physiology , src-Family Kinases/physiology , Adolescent , Adult , Amino Acid Sequence , Asthma/enzymology , Asthma/immunology , Asthma/metabolism , Eosinophils/metabolism , HL-60 Cells , Humans , Middle Aged , Molecular Sequence Data , Signal Transduction/immunology
9.
Biochem Biophys Res Commun ; 418(4): 669-75, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22293197

ABSTRACT

IL-3 is an important cytokine that regulates hematopoiesis. We have previously demonstrated that IL-3 is a potent inhibitor of osteoclastogenesis and bone resorption. In the present study, we have investigated the role of IL-3 on human osteoblast differentiation and bone formation. We found that IL-3 in a dose-dependent manner increases osteoblast differentiation and matrix mineralization in human mesenchymal stem cells (MSCs). IL-3 significantly enhances the expression of osteoblast specific genes such as alkaline phosphatase, collagen type-I, osteocalcin and osteopontin; and Runx-2 and osterix transcription factors. Moreover, IL-3 induces the expression of bone morphogenetic protein-2 (BMP-2), and activates smad1/5/8. IL-3 enhances osteoblast differentiation and BMP-2 secretion through JAK/STAT pathway. Interestingly, IL-3 promotes in vivo bone regeneration ability of MSCs. Thus, we reveal for the first time that IL-3 enhances human osteoblast differentiation and bone formation in both in vitro and in vivo conditions, and suggest its therapeutic potential for bone formation in important bone diseases.


Subject(s)
Cell Differentiation , Interleukin-3/physiology , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis , Alkaline Phosphatase/biosynthesis , Animals , Bone Morphogenetic Protein 2/biosynthesis , Bone Regeneration , Collagen Type I/biosynthesis , Core Binding Factor Alpha 1 Subunit/biosynthesis , Humans , Interleukin-3/pharmacology , Interleukin-3 Receptor alpha Subunit/biosynthesis , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Osteocalcin/biosynthesis , Osteopontin/biosynthesis , Sp7 Transcription Factor , Transcription Factors/biosynthesis
10.
Cell Immunol ; 280(1): 68-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23261831

ABSTRACT

Mast cells are the principal effectors of IgE-mediated immune responses, including allergic reactions. Tribbles homolog 3 (Trib3) encodes a pseudokinase implicated in the cellular stress response and has been linked to inflammation in certain situations. Here we report the role of Trib3 in mouse bone marrow-derived mast cells (BMMCs). Our results show that Trib3 mRNA expression in BMMCs is positively regulated by the growth factor interleukin (IL)-3. BMMCs originating from Trib3 knockout mice demonstrate unaltered differentiation kinetics and cell surface expression of mast cell markers. When challenged with transient IL-3 deprivation, Trib3-deficient BMMCs display delayed recovery, and during prolonged IL-3 starvation, cell death is accelerated in Trib3-null cultures. IgE-dependent and pharmacologically induced degranulation is impaired in Trib3-deficient BMMCs, as is activation-induced cytokine mRNA expression. Thus, Trib3 contributes to the survival and activity of primary cultured mast cells, which suggests a role for Trib3 in the modulation of the immune response.


Subject(s)
Cell Cycle Proteins/physiology , Gene Expression Regulation/drug effects , Interleukin-3/pharmacology , Mast Cells/drug effects , Animals , Bone Marrow Cells/cytology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cell Degranulation/drug effects , Cell Degranulation/physiology , Cell Differentiation/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , G1 Phase/drug effects , Immunoglobulin E/immunology , Interleukin-3/deficiency , Interleukin-3/physiology , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Passive Cutaneous Anaphylaxis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
11.
Cytokine ; 58(2): 267-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22370606

ABSTRACT

The crucial roles of the novel cytokine IL-33 in allergic, inflammatory, infectious and autoimmune diseases are becoming characterized. However, the cytokines which regulate IL-33 expression and secretion are still largely unknown. In this study, IL-3 and IL-4 were found to up-regulate IL-33 mRNA expression in mouse peritoneal exudate cells by a two-color DNA microarray and further confirmed by real time PCR and ELISA. IL-3 and IL-4 synergistically promote IL-33 mRNA expression and IL-33 intracrine in the heterogeneous cell populations as peritoneal exudates cells, bone marrow cells and splenic cells. IL-3 and IL-4 also induced IL-33 introcrine in the peritoneal exudate cells from the macrophage-deficient op/op mice, suggesting that macrophage is not the only target of IL-3 and IL-4 in the heterogeneous peritoneal exudate cells. Furthermore, IL-3 and IL-4 were verified to promote the IL-33 intracrine in the homogeneous cell population as fibroblasts and mast cells. These results indicate that up-regulation of IL-33 expression by IL-3 and IL-4 is not a feature particular to a specific type of cells. Up to 100 cytokines were screened, but none of them stimulated the secretion or release of IL-33 in the culture system. In summary, we confirm for the first time that IL-3 and IL-4 are critical for IL-33 intracrine in murine cells of various types, indicating that IL-3 and IL-4 may play an important role in the constitutive expression of IL-33 in vivo.


Subject(s)
Interleukin-3/physiology , Interleukin-4/physiology , Interleukins/metabolism , Up-Regulation , Animals , Base Sequence , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , Interleukin-33 , Ionomycin/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology
12.
J Immunol ; 184(3): 1143-7, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20038645

ABSTRACT

Basophils are recognized as immune modulators through their ability to produce IL-4, a key cytokine required for Th2 immunity. It has also recently been reported that basophils are transiently recruited into the draining lymph node (LN) after allergen immunization and that the recruited basophils promote the differentiation of naive CD4 T cells into Th2 effector cells. Using IL-3(-/-) and IL-3Rbeta(-/-) mice, we report in this study that the IL-3/IL-3R system is absolutely required to recruit circulating basophils into the draining LN following helminth infection. Unexpectedly, the absence of IL-3 or of basophil LN recruitment played little role in helminth-induced Th2 immune responses. Moreover, basophil depletion in infected mice did not diminish the development of IL-4-producing CD4 T cells. Our results reveal a previously unknown role of IL-3 in recruiting basophils to the LN and demonstrate that basophils are not necessarily associated with the development of Th2 immunity during parasite infection.


Subject(s)
Basophils/immunology , Basophils/pathology , Cell Movement/immunology , Interleukin-3/physiology , Lymph Nodes/immunology , Lymph Nodes/pathology , Strongylida Infections/immunology , Th2 Cells/immunology , Animals , Basophils/parasitology , Cell Movement/genetics , Gene Knock-In Techniques , Interleukin-3/deficiency , Interleukin-3/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nippostrongylus/immunology , Receptors, Interleukin-3/deficiency , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/physiology , Strongylida Infections/parasitology , Strongylida Infections/pathology , Th2 Cells/parasitology
13.
Dev Cell ; 11(2): 171-80, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16890157

ABSTRACT

Cytokines are important in adult hematopoiesis, yet their function in embryonic hematopoiesis has been largely unexplored. During development, hematopoietic stem cells (HSCs) are found in the aorta-gonad-mesonephros (AGM) region, yolk sac (YS), and placenta and require the Runx1 transcription factor for their normal generation. Since IL-3 is a Runx1 target and this cytokine acts on adult hematopoietic cells, we examined whether IL-3 affects HSCs in the mouse embryo. Using Runx1 haploinsufficient mice, we show that IL-3 amplifies HSCs from E11 AGM, YS, and placenta. Moreover, we show that IL-3 mutant embryos are deficient in HSCs and that IL-3 reveals the presence of HSCs in the AGM and YS prior to the stage at which HSCs are normally detected. Thus, our studies support an unexpected role for IL-3 during development and strongly suggest that IL-3 functions as a proliferation and/or survival factor for the earliest HSCs in the embryo.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Development , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Interleukin-3/physiology , Animals , Aorta/cytology , Cell Proliferation/drug effects , Cell Survival/drug effects , Core Binding Factor Alpha 2 Subunit/deficiency , Gonads/cytology , Hematopoietic Stem Cells/drug effects , Interleukin-3/genetics , Interleukin-3/pharmacology , Mesonephros/cytology , Mice , Placenta/cytology , Yolk Sac/cytology
14.
J Clin Invest ; 118(3): 924-34, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18246197

ABSTRACT

The proliferation and differentiation of hematopoietic stem cells (HSCs) is finely regulated by extrinsic and intrinsic factors via various signaling pathways. Here we have shown that, similar to mice deficient in the lipid phosphatase SHIP, loss of 2 Src family kinases, Lyn and Hck, profoundly affects HSC differentiation, producing hematopoietic progenitors with increased proliferation, reduced apoptosis, growth factor-independent survival, and skewed differentiation toward M2 macrophages. This phenotype culminates in a Stat5-dependent myeloproliferative disease that is accompanied by M2 macrophage infiltration of the lung. Expression of a membrane-bound form of SHIP in HSCs lacking both Lyn and Hck restored normal hematopoiesis and prevented myeloproliferation. In vitro and in vivo studies suggested the involvement of autocrine and/or paracrine production of IL-3 and GM-CSF in the increased proliferation and myeloid differentiation of HSCs. Thus, this study has defined a myeloproliferative transformation-sensitive signaling pathway, composed of Lyn/Hck, SHIP, autocrine/paracrine cytokines, and Stat5, that regulates HSC differentiation and M2 macrophage programming.


Subject(s)
Hematopoietic Stem Cells/cytology , Macrophages/cytology , Phosphoric Monoester Hydrolases/physiology , Proto-Oncogene Proteins c-hck/physiology , STAT5 Transcription Factor/physiology , src-Family Kinases/physiology , Animals , Cell Differentiation , Cell Proliferation , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Inositol Polyphosphate 5-Phosphatases , Interleukin-3/physiology , Lung/pathology , Lung Diseases/etiology , Mice , Mice, Knockout , Myeloproliferative Disorders/etiology , Signal Transduction
15.
Blood ; 113(26): 6658-68, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19282460

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a potent adjuvant in cancer vaccination; however, the specific role of endogenous GM-CSF remains unknown. We performed cell-based vaccination in 2 tumor models. First, we vaccinated C57BL/6 mice lacking either GM-CSF, IL-5, or beta-common chain (betac), a receptor subunit essential for GM-CSF and IL-5 signaling, with melanoma cells engineered to produce GM-CSF. Tumor vaccination was effective in both GM-CSF(-/-) and IL-5(-/-) mice, showing that protective immunization is independent of both endogenous cytokines. However, all betac(-/-) animals developed tumor. Loss of tumor immunity in betac(-/-) mice does not reflect global impairment in cell-mediated immunity, as contact hypersensitivity reaction to haptens is unaltered. The importance of tumor cell-derived GM-CSF was highlighted by recruitment of dendritic cells at the vaccination site in wild-type, GM-CSF(-/-), and IL-5(-/-) but not in betac(-/-) mice. In the second model, vaccination with unmodified RENCA cells showed similar results with efficient immunization in BALB/c wild-type and GM-CSF(-/-), whereas all betac(-/-) animals died. Altogether, our results strongly suggest that although endogenous GM-CSF and IL-5 are not required to induce tumor immunity, signaling through betac receptor is critically needed for efficient cancer vaccination in both genetically modified GM-CSF-secreting tumor cells and a spontaneously immunogenic models.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Renal Cell/prevention & control , Cytokine Receptor Common beta Subunit/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Melanoma, Experimental/prevention & control , Animals , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor/metabolism , Cell Line, Tumor/transplantation , Culture Media, Conditioned/chemistry , Cytokine Receptor Common beta Subunit/deficiency , Cytokine Receptor Common beta Subunit/genetics , Cytokines/analysis , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Injections, Subcutaneous , Interleukin-3/deficiency , Interleukin-3/genetics , Interleukin-3/physiology , Interleukin-5/deficiency , Interleukin-5/genetics , Interleukin-5/physiology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins/physiology , Species Specificity , Vaccination/methods
16.
J Immunol ; 182(2): 1050-60, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19124748

ABSTRACT

IL-3 plays important roles in the growth and survival of hematopoietic progenitor cells, processes modeled in studies of the IL-3-dependent cell line Ba/F3. To gain insights into molecular mechanisms governing cell fate, we examined the patterns of proteins up-regulated following stimulation of Ba/F3 cells with IL-3. Through two-dimensional electrophoresis and proteomics-based approaches, we identified 11 proteins. Of these, expression of 14-3-3gamma was significantly increased by IL-3 stimulation at both the transcriptional and translational levels. 14-3-3gamma overexpression in Ba/F3 cells abrogated dependence on IL-3 and was associated with activation of PI3K and MAPK signaling cascades, suggesting that the functions of 14-3-3gamma in normal hematopoietic progenitors are to promote survival and growth through the activation of distinct signaling pathways. Additionally, the up-regulation of Bax and Bad was seen with the ablation of 14-3-3gamma, resulting in cell death. These results indicate that deregulated expression of 14-3-3gamma may contribute to malignant transformation, possibly providing a new target for therapeutic intervention in hematopoietic neoplasms.


Subject(s)
14-3-3 Proteins/physiology , Cell Proliferation , Interleukin-3/physiology , 14-3-3 Proteins/biosynthesis , 14-3-3 Proteins/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Gene Expression Profiling , Humans , K562 Cells , Leukemia L1210/genetics , Leukemia L1210/metabolism , Leukemia L1210/pathology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , Proteomics/methods , Signal Transduction/genetics , Signal Transduction/immunology
17.
J Immunol ; 182(1): 361-70, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109167

ABSTRACT

IL-3, a cytokine secreted by activated T cells is well known to regulate the proliferation, differentiation, and survival of pluripotent hematopoietic stem cells. IL-3 functions as a link between the immune and the hematopoietic system. In this study, we suggest an important new role of IL-3 in inhibition of TNF-alpha-induced bone resorption in vitro and prevention of inflammatory arthritis in mice. We show here that IL-3 potently and irreversibly inhibits TNF-alpha-induced bone resorption in hematopoietic precursors of monocyte/macrophage lineage. IL-3 showed an inhibitory effect on TNF-alpha-induced bone resorption even in the presence of proinflammatory cytokines such as IL-1alpha, TGF-beta(1), TGF-beta(3), IL-6, and PGE(2). We found that IL-3 prevented TNF-alpha-induced c-fos nuclear translocation and AP-1 DNA-binding activity. Interestingly, IL-3 pretreatment prevented the development of inflammatory arthritis in mice induced by a mixture of anti-type II collagen mAbs and LPS. Furthermore, IL-3 prevented cartilage and bone loss in the joints indirectly through inhibition of inflammation. Thus, we provide the first evidence that IL-3, a strong regulator of hematopoiesis, also plays an important role in inhibition of TNF-alpha-induced bone resorption and prevention of inflammatory arthritis in mice.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Bone Resorption/immunology , Bone Resorption/prevention & control , Inflammation Mediators/physiology , Interleukin-3/physiology , Tumor Necrosis Factor-alpha/physiology , Active Transport, Cell Nucleus/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/metabolism , Bone Resorption/pathology , Cartilage, Articular/immunology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cells, Cultured , Collagen Type II/immunology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Inflammation Mediators/administration & dosage , Interleukin-3/administration & dosage , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Osteochondritis/immunology , Osteochondritis/metabolism , Osteochondritis/prevention & control , Protein Binding/genetics , Protein Binding/immunology , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors
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.
J Biosci ; 462021.
Article in English | MEDLINE | ID: mdl-34183475

ABSTRACT

IL-3, a haematopoiesis regulatory factor, has previously been shown to inhibit both mouse and human osteoclast differentiation and bone resorption. Here, the role of rat IL-3 on rat osteoclast differentiation was evaluated to address whether the inhibitory action of IL-3 on osteoclastogenesis is conserved in various species. It was observed that IL-3 inhibited rat osteoclast differentiation induced by both TNF-α and receptor activator of NF-ĸB ligand (RANKL). TNF-α is known to induce bone loss in postmenopausal osteoporotic women and it also synergise with many pro-osteoclastogenic cytokines to cause huge pathological bone loss. Importantly, it was found that rat IL-3 inhibits the synergistic action of TNF-α with RANKL and IL-1ß, TGFß1 and TGF-ß3. IL-3 downregulates the TNF-α-induced nuclear translocation of NF-ĸB-p65 and c-fos without affecting c-jun. Interestingly, we observed that IL-3 also inhibits osteoclast differentiation in vivo in rats induced by TNF-α. All these results suggest that inhibitory action of IL-3 on osteoclastogenesis is conserved in various species including mice, rats and humans. Thus, our results clearly indicate that IL-3 has therapeutic potential to treat pathological bone loss in important skeletal diseases.


Subject(s)
Cell Differentiation , Interleukin-3/physiology , Osteoclasts , Osteogenesis , Animals , NF-kappa B/metabolism , RANK Ligand/metabolism , Rats, Wistar , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
J Exp Med ; 173(2): 507-10, 1991 Feb 01.
Article in English | MEDLINE | ID: mdl-1899106

ABSTRACT

We have characterized the mast cell stimulating activity of murine cytokine synthesis inhibitory factor, referred to as interleukin 10 (IL-10). It was found that IL-10 alone failed to support the growth of mast cell lines and mast cell progenitors. Nevertheless, it dramatically enhanced their growth when combined with IL-3 or IL-4. Moreover, IL-4 plus IL-10 supported the proliferation of mast cells as well as IL-3, suggesting that these two factors may provide a pathway for their development independent of IL-3. However, optimal mast cell growth was stimulated by the combination of IL-10, IL-4, and IL-3. This particular set of cytokines are coordinately produced by activated T cells and may constitute an effective network regulating early and late stages of mast cell development during certain immune responses.


Subject(s)
Interleukins/physiology , Mast Cells/cytology , Animals , Cell Division , Cell Line , Interleukin-10 , Interleukin-3/physiology , Interleukin-4/physiology , Mast Cells/ultrastructure , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Strains , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL