Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 618
Filter
1.
Front Immunol ; 15: 1411047, 2024.
Article in English | MEDLINE | ID: mdl-38938573

ABSTRACT

Interleukin (IL)-3 has long been known for its hematopoietic properties. However, recent evidence has expanded our understanding of IL-3 function by identifying IL-3 as a critical orchestrator of inflammation in a wide array of diseases. Depending on the type of disease, the course of inflammation, the cell or the tissue involved, IL-3 promotes either pathologic inflammation or its resolution. Here, we describe the cell-specific functions of IL-3 and summarize its role in diseases. We discuss the current treatments targeting IL-3 or its receptor, and highlight the potential and the limitations of targeting IL-3 in clinics.


Subject(s)
Inflammation , Interleukin-3 , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-3/metabolism , Animals , Signal Transduction , Receptors, Interleukin-3/metabolism , Receptors, Interleukin-3/immunology
2.
Cell Commun Signal ; 22(1): 254, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702781

ABSTRACT

IL-3/STAT5 signaling pathway is crucial for the development and activation of immune cells, contributing to the cellular response to infections and inflammatory stimuli. Dysregulation of the IL-3/STAT5 signaling have been associated with inflammatory and autoimmune diseases characterized by inflammatory cell infiltration and organ damage. IL-3 receptor α (IL-3Rα) specifically binds to IL-3 and initiates intracellular signaling, resulting in the phosphorylation of STAT5. However, the regulatory mechanisms of IL-3Rα remain unclear. Here, we identified the E3 ubiquitin ligase RNF128 as a negative regulator of IL-3/STAT5 signaling by targeting IL-3Rα for lysosomal degradation. RNF128 was shown to selectively bind to IL-3Rα, without interacting with the common beta chain IL-3Rß, which shares the subunit with GM-CSF. The deficiency of Rnf128 had no effect on GM-CSF-induced phosphorylation of Stat5, but it resulted in heightened Il-3-triggered activation of Stat5 and increased transcription of the Id1, Pim1, and Cd69 genes. Furthermore, we found that RNF128 promoted the K27-linked polyubiquitination of IL-3Rα in a ligase activity-dependent manner, ultimately facilitating its degradation through the lysosomal pathway. RNF128 inhibited the activation and chemotaxis of macrophages in response to LPS stimulation, thereby attenuating excessive inflammatory responses. Collectively, these results reveal that RNF128 negatively regulates the IL-3/STAT5 signaling pathway by facilitating K27-linked polyubiquitination of IL-3Rα. This study uncovers E3 ubiquitin ligase RNF128 as a novel regulator of the IL-3/STAT5 signaling pathway, providing potential molecular targets for the treatment of inflammatory diseases.


Subject(s)
Interleukin-3 , STAT5 Transcription Factor , Signal Transduction , Ubiquitin-Protein Ligases , Ubiquitination , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Humans , Animals , Interleukin-3/metabolism , Mice , Lysosomes/metabolism , HEK293 Cells , Phosphorylation , Receptors, Interleukin-3/metabolism , Receptors, Interleukin-3/genetics
3.
Neurochem Res ; 49(5): 1373-1386, 2024 May.
Article in English | MEDLINE | ID: mdl-38512424

ABSTRACT

Interleukin 3 (IL-3) is a well-known pleiotropic cytokine that regulates the proliferation and differentiation of hematopoietic progenitor cells, triggering classical signaling pathways such as JAK/STAT, Ras/MAPK, and PI3K/Akt to carry out its functions. Interestingly, the IL-3 receptor is also expressed in non-hematopoietic cells, playing a crucial role in cell survival. Our previous research demonstrated the expression of the IL-3 receptor in neuron cells and its protective role in neurodegeneration. Glutamate, a principal neurotransmitter in the central nervous system, can induce cellular stress and lead to neurotoxicity when its extracellular concentrations surpass normal levels. This excessive glutamate presence is frequently observed in various neurological diseases. In this study, we uncover the protective role of IL-3 as an inhibitor of glutamate-induced cell death, analyzing the cytokine's signaling pathways during its protective effect. Specifically, we examined the relevance of JAK/STAT, Ras/MAPK, and PI3 K signaling pathways in the molecular mechanism triggered by IL-3. Our results show that the inhibition of JAK, ERK, and PI3 K signaling pathways, using pharmacological inhibitors, effectively blocked IL-3's protective role against glutamate-induced cell death. Additionally, our findings suggest that Bcl-2 and Bax proteins may be involved in the molecular mechanism triggered by IL-3. Our investigation into IL-3's ability to protect neuronal cells from glutamate-induced damage offers a promising therapeutic avenue with potential clinical implications for several neurological diseases characterized by glutamate neurotoxicity.


Subject(s)
Interleukin-3 , Neuroblastoma , Humans , Glutamic Acid/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Interleukin-3 , Cell Line , Proto-Oncogene Proteins c-akt/metabolism
4.
Gut ; 72(11): 2081-2094, 2023 11.
Article in English | MEDLINE | ID: mdl-37541770

ABSTRACT

IL-3 has been reported to be involved in various inflammatory disorders, but its role in inflammatory bowel disease (IBD) has not been addressed so far. Here, we determined IL-3 expression in samples from patients with IBD and studied the impact of Il3 or Il3r deficiency on T cell-dependent experimental colitis. We explored the mechanical, cytoskeletal and migratory properties of Il3r -/- and Il3r +/+ T cells using real-time deformability cytometry, atomic force microscopy, scanning electron microscopy, fluorescence recovery after photobleaching and in vitro and in vivo cell trafficking assays. We observed that, in patients with IBD, the levels of IL-3 in the inflamed mucosa were increased. In vivo, experimental chronic colitis on T cell transfer was exacerbated in the absence of Il-3 or Il-3r signalling. This was attributable to Il-3r signalling-induced changes in kinase phosphorylation and actin cytoskeleton structure, resulting in increased mechanical deformability and enhanced egress of Tregs from the inflamed colon mucosa. Similarly, IL-3 controlled mechanobiology in human Tregs and was associated with increased mucosal Treg abundance in patients with IBD. Collectively, our data reveal that IL-3 signaling exerts an important regulatory role at the interface of biophysical and migratory T cell features in intestinal inflammation and suggest that this might be an interesting target for future intervention.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , T-Lymphocytes, Regulatory , Receptors, Interleukin-3/metabolism , Interleukin-3/metabolism , Inflammation/metabolism , Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism
5.
Asian Pac J Cancer Prev ; 24(1): 185-194, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36708567

ABSTRACT

BACKGROUND: Quantitative profiling of specific cell surface markers is a new approach in characterization of tumor heterogeneity and single cell biology. The current tools have dearth in detection and quantification of receptor proteins on single cells. METHODS: we focused on our newly developed protocol to determine the distribution pattern and density of cell surface markers on single acute myeloid leukemia cells. Cell surface proteins were labeled with quantum dots (Qdots) followed by super resolution Structured Illumination Microscopy (SIM) imaging to imprisonment the optical signals emitted by Qdots which were further analyzed by software imaris to do three dimensional (3D) structure reconstruction and digital simulation. Furthermore, MTT assays and flow cytometry was performed to establish association between expression of cell surface markers and drug response. RESULTS: In the present study, we found that the Molm13 and cytarabine-enriched Molm13 cells exhibit different densities of CD123, an alpha-subunit of interleukin-3 receptor, i.e. 0.92 and 1.73 per µm2 of cell surface respectively. Sub-populations of Molm13 cells expressing higher densities of CD123 on cells membranes showed resistance against cytarabine. Further study revealed that romidepsin sensitized and augmented the cytotoxicity of cytarabine in Molm13 and cytarabine-enriched Molm13 cells. Romidepsin increased the percentage of cell death-induced by cytarabine from 21.6 % to 28.6 % and 37.1 % to 57.2 % in Molm13 and cytarabine-enriched Molm13 cells respectively. CONCLUSION: Altogether, the study suggests that Molm13 cells have sub-populations with differential expression of CD123+ phenotype. Romidepsin sensitizes and augments the effect of cytarabine in Molm13 and cytarabine-enriched Molm13 cells.


Subject(s)
Leukemia, Myeloid, Acute , Receptors, Interleukin-3 , Humans , Interleukin-3 Receptor alpha Subunit , Leukemia, Myeloid, Acute/pathology , Cytarabine/pharmacology , Flow Cytometry , Cell Line, Tumor
6.
Cell Immunol ; 374: 104498, 2022 04.
Article in English | MEDLINE | ID: mdl-35334276

ABSTRACT

Basophils and mast cells play a critical role in allergic inflammation and provide protective immunity against certain types of parasitic infections. Expansion of basophils and mast cells to the critical numbers is believed to be an essential step in enabling basophils and mast cells to carry out their protective functions. However, factors that drive basophil and mast cell expansion are still incompletely understood. We tested the roles of cytokines and growth factors IL-3, TSLP, GM-CSF, IL-5, SCF, IL-7, IL-25, and IL-33 in promoting the differentiation of pre-basophil and mast cell progenitors (pre-BMPs)in vitro.We found that while GM-CSF only expanded basophils, IL-3 promoted the differentiation of pre-BMPs into both basophils and mast cells. We found that IL-3 expanded the number of pre-BMPsin vivo. We showed that IL-3 upregulatedIl3ramRNA and protein expression on pre-BMPs, supporting that IL-3 expands pre-BMPs in part by upregulating the IL-3 receptor expression. Although Gata2 mRNA expression was upregulated by IL-3 treatment in pre-BMPs, it is dispensable for IL-3-mediated upregulation of IL-3 receptor expression. Our study reveals a novel mechanism through which IL-3 expands basophil and mast cells.


Subject(s)
Basophils , Receptors, Interleukin-3 , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-3 , Mast Cells
7.
Cell Death Dis ; 13(2): 137, 2022 02 10.
Article in English | MEDLINE | ID: mdl-35145069

ABSTRACT

Acute respiratory distress syndrome (ARDS) is triggered by various aetiological factors such as trauma, sepsis and respiratory viruses including SARS-CoV-2 and influenza A virus. Immune profiling of severe COVID-19 patients has identified a complex pattern of cytokines including granulocyte macrophage-colony stimulating factor (GM-CSF) and interleukin (IL)-5, which are significant mediators of viral-induced hyperinflammation. This strong response has prompted the development of therapies that block GM-CSF and other cytokines individually to limit inflammation related pathology. The common cytokine binding site of the human common beta (ßc) receptor signals for three inflammatory cytokines: GM-CSF, IL-5 and IL-3. In this study, ßc was targeted with the monoclonal antibody (mAb) CSL311 in engineered mice devoid of mouse ßc and ßIL-3 and expressing human ßc (hßcTg mice). Direct pulmonary administration of lipopolysaccharide (LPS) caused ARDS-like lung injury, and CSL311 markedly reduced lung inflammation and oedema, resulting in improved oxygen saturation levels in hßcTg mice. In a separate model, influenza (HKx31) lung infection caused viral pneumonia associated with a large influx of myeloid cells into the lungs of hßcTg mice. The therapeutic application of CSL311 potently decreased accumulation of monocytes/macrophages, neutrophils, and eosinophils without altering lung viral loads. Furthermore, CSL311 treatment did not limit the viral-induced expansion of NK and NKT cells, or the tissue expression of type I/II/III interferons needed for efficient viral clearance. Simultaneously blocking GM-CSF, IL-5 and IL-3 signalling with CSL311 may represent an improved and clinically applicable strategy to reducing hyperinflammation in the ARDS setting.


Subject(s)
Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/physiology , Respiratory Distress Syndrome/immunology , Animals , Antibodies, Monoclonal/immunology , Cytokine Receptor Common beta Subunit/immunology , Cytokines , Eosinophils/immunology , Female , Humans , Immunity/genetics , Immunity/physiology , Inflammation/immunology , Leukocytes/metabolism , Male , Mice , Mice, Transgenic , Neutrophils/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Receptors, Interleukin-3 , Receptors, Interleukin-5 , Respiratory Distress Syndrome/physiopathology
8.
Amino Acids ; 53(8): 1211-1227, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34196789

ABSTRACT

Interleukin-3 (IL-3) is a cytokine belonging to the family of common ß (ßc) and is involved in various biological systems. Its activity is mediated by the interaction with its receptor (IL-3R), a heterodimer composed of two distinct subunits: IL-3Rα and ßc. IL-3 and its receptor, especially IL-3Rα, play a crucial role in pathologies like inflammatory diseases and therefore are interesting therapeutic targets. Here, we have performed an analysis of these proteins and their interaction based on structural and evolutionary information. We highlighted that IL-3 and IL-3Rα structural architectures are conserved across evolution and shared with other proteins belonging to the same ßc family interleukin-5 (IL-5) and granulocyte-macrophage colony-stimulating factor (GM-CSF). The IL-3Rα/IL-3 interaction is mediated by a large interface in which most residues are surprisingly not conserved during evolution and across family members. In spite of this high variability, we suggested small regions constituted by few residues conserved during the evolution in both proteins that could be important for the binding affinity.


Subject(s)
Evolution, Molecular , Interleukin-3/chemistry , Receptors, Interleukin-3/chemistry , Amino Acid Sequence , Animals , Humans , Interleukin-3/genetics , Protein Conformation , Receptors, Interleukin-3/genetics , Sequence Homology, Amino Acid
9.
Cells ; 9(9)2020 09 17.
Article in English | MEDLINE | ID: mdl-32957735

ABSTRACT

Bone marrow-derived mast cells (BMMCs) are often used as a model system for studies of the role of MCs in health and disease. These cells are relatively easy to obtain from total bone marrow cells by culturing under the influence of IL-3 or stem cell factor (SCF). After 3 to 4 weeks in culture, a nearly homogenous cell population of toluidine blue-positive cells are often obtained. However, the question is how relevant equivalents these cells are to normal tissue MCs. By comparing the total transcriptome of purified peritoneal MCs with BMMCs, here we obtained a comparative view of these cells. We found several important transcripts that were expressed at very high levels in peritoneal MCs, but were almost totally absent from the BMMCs, including the major chymotryptic granule protease Mcpt4, the neurotrophin receptor Gfra2, the substance P receptor Mrgprb2, the metalloprotease Adamts9 and the complement factor 2 (C2). In addition, there were a number of other molecules that were expressed at much higher levels in peritoneal MCs than in BMMCs, including the transcription factors Myb and Meis2, the MilR1 (Allergin), Hdc (Histidine decarboxylase), Tarm1 and the IL-3 receptor alpha chain. We also found many transcripts that were highly expressed in BMMCs but were absent or expressed at low levels in the peritoneal MCs. However, there were also numerous MC-related transcripts that were expressed at similar levels in the two populations of cells, but almost absent in peritoneal macrophages and B cells. These results reveal that the transcriptome of BMMCs shows many similarities, but also many differences to that of tissue MCs. BMMCs can thereby serve as suitable models in many settings concerning the biology of MCs, but our findings also emphasize that great care should be taken when extrapolating findings from BMMCs to the in vivo function of tissue-resident MCs.


Subject(s)
B-Lymphocytes/metabolism , Bone Marrow Cells/metabolism , Macrophages/metabolism , Mast Cells/metabolism , Peritoneum/metabolism , Transcriptome , ADAMTS9 Protein/genetics , ADAMTS9 Protein/metabolism , Animals , B-Lymphocytes/cytology , Biomarkers/metabolism , Bone Marrow Cells/cytology , Complement C2/genetics , Complement C2/metabolism , Female , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Macrophages/cytology , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Organ Specificity , Peritoneum/cytology , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
10.
Hum Vaccin Immunother ; 16(10): 2341-2348, 2020 10 02.
Article in English | MEDLINE | ID: mdl-32692611

ABSTRACT

The IL-3 alpha chain receptor (CD123) is a cell surface protein that is widely expressed by various subtypes of acute leukemia, including acute myeloid leukemia (AML), acute lymphoblastic leukemia and blastic plasmacytoid dendritic cell neoplasm. Notably, CD123 is preferentially overexpressed in leukemia stem cells (LSC) in contrast to normal hematopoietic stem cells, and this differential expression allows for the selective eradication of LSC and leukemic blasts through therapeutic targeting of CD123, with less impact on hematopoietic cells. The level of CD123 expression in AML correlates with both treatment response and outcomes. Therefore, targeting CD123 represents a promising universal therapeutic target in advanced acute leukemias irrespective of the individual leukemia phenotype. There are currently 31 ongoing clinical trials examining the utility of CD123-based targeted therapies. Here we focus our review on current efforts to target CD123 in acute leukemia through various therapeutic constructs.


Subject(s)
Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Interleukin-3 Receptor alpha Subunit , Leukemia, Myeloid, Acute/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Interleukin-3
11.
Immunology ; 159(4): 441-449, 2020 04.
Article in English | MEDLINE | ID: mdl-31957000

ABSTRACT

Signaling by Kit has been extensively studied in hematopoietic cells and is essential for the survival, proliferation and maintenance of hematopoietic stem and progenitor cells. In addition to the activation of intrinsic signaling pathways, Kit has been shown to interact with lineage-restricted type I cytokine receptors and produce cross signals, e.g. erythropoietin receptor, interleukin-7 receptor (IL-7R), IL-3R. Based on the earlier studies, we hypothesize that Kit activate other type I cytokine receptors in a cell-specific manner and execute cell-specific function. To investigate other Kit-activated receptors, we tested Kit and IL-4R cross-receptor activation in murine bone-marrow-derived mast cells, which express both Kit and IL-4R at the surface level. Kit upon activation by Kit ligand (KL), activated IL-4Rα, γC , and signal transducer and activator of transcription 6 independent of its cognate ligand IL-4. Though KL and IL-4 are individually mitogenic, combinations of KL and IL-4 synergistically promoted mast cell proliferation. Furthermore, inhibition of lipid raft formation by methyl-ß-cyclodextrin resulted in loss of synergistic proliferation. Together the data suggest IL-4R as a novel Kit-activated receptor. Such cross-receptor activations are likely to be a universal mechanism of Kit signaling in hematopoiesis.


Subject(s)
Interleukin-4/pharmacology , Mast Cells/drug effects , Proto-Oncogene Proteins c-kit/genetics , Receptors, Interleukin-4/genetics , STAT6 Transcription Factor/genetics , Stem Cell Factor/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Proliferation/drug effects , Gene Expression Regulation , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoiesis/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mast Cells/cytology , Mast Cells/immunology , Membrane Microdomains/drug effects , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Primary Cell Culture , Proto-Oncogene Proteins c-kit/immunology , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/immunology , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Receptors, Interleukin-4/immunology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , STAT6 Transcription Factor/immunology , Signal Transduction , Stem Cell Factor/genetics , Stem Cell Factor/immunology , beta-Cyclodextrins/pharmacology
12.
J Immunol ; 204(4): 819-831, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31900339

ABSTRACT

IL-3, a cytokine secreted by activated T lymphocytes, is known to regulate the proliferation, survival, and differentiation of hematopoietic cells. However, the role of IL-3 in regulation of T cell functions is not fully delineated. Previously, we have reported that IL-3 plays an important role in development of regulatory T cells in mice. In this study, we investigated the regulation of IL-3R expression on human Th cells and also examined the role of IL-3 in effector functions of these cells. We found that human peripheral blood Th cells in resting state do not show surface expression of IL-3R; however, its expression was observed at transcript and intracellular protein levels. The functional IL-3R expression on the surface was seen only after antigenic stimulation. When naive Th cells were activated in the presence of various cytokines, we found that IL-4 significantly increases the surface expression of IL-3R and also increases the number of IL-3R+ Th cells. Interestingly, IL-3R+ cells exhibit a Th2 cell-like phenotype and show high GATA-3 expression. Moreover, Th2 cells in presence of IL-3 show increased expression of type 2 effector cytokines, such as IL-4, IL-5, and IL-13. Furthermore, IL-3R expressing and IL-3-secreting Th cells were high in house dust mite-allergic patients. Thus, to our knowledge, we provide the first evidence that the expression of IL-3R on activated human Th cells is modulated by IL-4, and IL-3 regulates the effector functions of Th2 cells. Our results suggest that IL-3 may play an important role in regulating allergic immune responses.


Subject(s)
Cell Differentiation/immunology , Interleukin-3/immunology , Interleukin-4/immunology , Receptors, Interleukin-3/immunology , Th2 Cells/immunology , Humans , Hypersensitivity/immunology , Interleukin-3/metabolism , Interleukin-4/metabolism , Lymphocyte Activation/immunology , Receptors, Interleukin-3/metabolism
13.
Immunity ; 50(4): 796-811, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995500

ABSTRACT

The ß common chain cytokines GM-CSF, IL-3, and IL-5 regulate varied inflammatory responses that promote the rapid clearance of pathogens but also contribute to pathology in chronic inflammation. Therapeutic interventions manipulating these cytokines are approved for use in some cancers as well as allergic and autoimmune disease, and others show promising early clinical activity. These approaches are based on our understanding of the inflammatory roles of these cytokines; however, GM-CSF also participates in the resolution of inflammation, and IL-3 and IL-5 may also have such properties. Here, we review the functions of the ß common cytokines in health and disease. We discuss preclinical and clinical data, highlighting the potential inherent in targeting these cytokine pathways, the limitations, and the important gaps in understanding of the basic biology of this cytokine family.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Inflammation/immunology , Interleukin-3/immunology , Interleukin-5/immunology , Animals , Autoimmune Diseases/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Hematopoiesis/immunology , Humans , Inflammation/therapy , Interleukin-3/antagonists & inhibitors , Interleukin-3/deficiency , Interleukin-3/genetics , Interleukin-5/antagonists & inhibitors , Interleukin-5/deficiency , Interleukin-5/genetics , Mice , Mice, Knockout , Multigene Family , Neoplasms/immunology , Neoplasms/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Receptors, Interleukin-5/genetics , Receptors, Interleukin-5/immunology , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Signal Transduction , Structure-Activity Relationship , Vaccination , Wound Healing/immunology
14.
Mol Immunol ; 109: 71-80, 2019 05.
Article in English | MEDLINE | ID: mdl-30870654

ABSTRACT

Sepsis is defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. There are multiple cytokines involved in the process of sepsis. As an important upstream cytokine in inflammation, Interleukin-3 (IL-3) plays a crucial role during sepsis, however, its exact role is unclear. The purpose of this study is to discuss the role of IL-3 and its receptor in cecal ligation and puncture (CLP)-induced sepsis in a rat model. The Cluster of Differentiation 123 (CD123, IL-3 receptor alpha chain, IL-3Rac) antibody (anti-CD123) was used to directly target IL-3's receptor and alleviate the effect of IL-3 in the CLP + anti-CD123 group during the early stage of sepsis. CLP was performed in the CLP and CLP + anti-CD123 groups. The time points of observation included 12 h, 24 h, and 5d after the operation. The results showed that the rats in the CLP + anti-CD123 group had lower levels of lactate, serum tumor necrosis factor-α (TNF-α), Interleukin-1ß (IL-1ß), and Interleukin-6 (IL-6), and also exhibited a higher core temperature, mean arterial pressure (MAP), Oxygenation Index (PO2/FiO2), and end-tidal carbon dioxide (ETCO2) and serum Interleukin-10 (IL-10) levels after CLP than those in the CLP group. Additionally, administration of anti-CD123 led to a stable down-regulation of tyrosine phosphorylation of the IL-3 receptor, a decline in phosphorylation of the Janus kinase 2 (JAK2) protein, and the signal transduction and activation of transcription 5 (STAT5) proteins in lung tissues. Meanwhile, the study revealed that treatment of anti-CD123 can markedly attenuate histological damages in lung and kidney tissues, improve sublingual microcirculation, and prolong survival post sepsis. In conclusion, anti-CD123 reduces mortality and alleviates organ dysfunction by restraining the JAK2-STAT5 signaling pathway and reduces serum cytokines in the development of early sepsis in a rat model induced by CLP.


Subject(s)
Cecum/pathology , Receptors, Interleukin-3/antagonists & inhibitors , Sepsis/pathology , Sepsis/prevention & control , Animals , Antibodies/pharmacology , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-3/metabolism , Interleukin-3 Receptor alpha Subunit/metabolism , Kidney/drug effects , Kidney/pathology , Ligation , Lung/metabolism , Lung/pathology , Male , Microcirculation/drug effects , Punctures , Rats, Sprague-Dawley , Receptors, Interleukin-3/metabolism , Signal Transduction/drug effects
15.
Cell Immunol ; 334: 49-60, 2018 12.
Article in English | MEDLINE | ID: mdl-30274838

ABSTRACT

A large number of animal models revealed that IL-3 plays an important role for the development of T and B cell-mediated autoimmune diseases. However, little is known about the expression and regulation of IL-3 receptors in human T and B cells and how IL-3 modulates the activation and survival of these cells. We show that the IL-3 receptor CD123 is substantially upregulated on proliferating CD4+ and CD8+ T as well as B cells. Upregulation of CD123 differs between various activators and can be further modulated by cytokines. Exposure of human T and B cells to IL-3 enhances proliferation and survival. IL-3 also induces a shift towards secretion of proinflammatory cytokines in T and B cells and reduces the expression of IL-10 in B cells. Thus IL-3 may have proinflammatory and immunostimulatory properties also in human autoimmune diseases.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-3/immunology , Receptors, Interleukin-3/immunology , Autoimmune Diseases/immunology , Cytokines/immunology , Humans , Interleukin-3 Receptor alpha Subunit/immunology , Lymphocyte Activation/immunology , Up-Regulation/immunology
16.
Theranostics ; 7(5): 1333-1345, 2017.
Article in English | MEDLINE | ID: mdl-28435469

ABSTRACT

Despite Imatinib (IM), a selective inhibitor of Bcr-Abl, having led to improved prognosis in Chronic Myeloid Leukemia (CML) patients, acquired resistance and long-term adverse effects is still being encountered. There is, therefore, urgent need to develop alternative strategies to overcome drug resistance. According to the molecules expressed on their surface, exosomes can target specific cells. Exosomes can also be loaded with a variety of molecules, thereby acting as a vehicle for the delivery of therapeutic agents. In this study, we engineered HEK293T cells to express the exosomal protein Lamp2b, fused to a fragment of Interleukin 3 (IL3). The IL3 receptor (IL3-R) is overexpressed in CML blasts compared to normal hematopoietic cells and thus is able to act as a receptor target in a cancer drug delivery system. Here we show that IL3L exosomes, loaded with Imatinib or with BCR-ABL siRNA, are able to target CML cells and inhibit in vitro and in vivo cancer cell growth.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cell Proliferation/drug effects , Drug Carriers/metabolism , Exosomes/metabolism , Imatinib Mesylate/pharmacokinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Receptors, Interleukin-3/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Heterografts , Humans , Imatinib Mesylate/administration & dosage , Mice , Treatment Outcome
17.
Mol Pharm ; 14(3): 940-952, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28146632

ABSTRACT

Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults and is associated with high relapse rates. It is known that leukemia stem cells (LSCs), a very small subpopulation of the total number of leukemic cells, maintain the leukemia phenotype (∼80-90% of AML remain the same as at first diagnosis), display chemotherapy resistance, and contribute to disease regeneration. Therefore, targeting LSCs could control the relapse of AML. Small interfering RNA (siRNA), an effector of the RNA interference (RNAi) pathway, can selectively downregulate any gene implicated in the pathology of disease, presenting great potential for treatment of AML. In this study an antibody targeted cyclodextrin-based nanoparticle (NP) (CD.DSPE-PEG-Fab) was developed for siRNA delivery specifically to AML LSCs. The targeted CD.siRNA.DSPE-PEG-Fab formulation, where Fab specifically targets the IL-3 receptor α-chain (IL-3Rα, also known as CD123, a cell surface antigen for human AML LSCs), achieved antigen-mediated cellular uptake in KG1 cells (an AML leukemia stem and progenitor cell line). Efficient delivery of bromodomain-containing protein 4 (BRD4) siRNA using the targeted formulation resulted in downregulation of the corresponding mRNA and protein in KG1 cells and in ex vivo primary AML patient derived samples. The resulting silencing of BRD4 induced myeloid differentiation and triggered leukemia apoptosis. In addition, a synergistic therapeutic effect was detected when administered in combination with the chemotherapeutic, cytarabine (Ara-C). These results indicate the clinical potential of the antibody-tagged cyclodextrin NP for targeted delivery of therapeutic siRNA in the treatment of AML.


Subject(s)
Antibodies/administration & dosage , Cyclodextrins/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cytarabine/pharmacology , Down-Regulation/drug effects , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Receptors, Interleukin-3/metabolism , Transcription Factors/metabolism
18.
Leukemia ; 31(4): 957-966, 2017 04.
Article in English | MEDLINE | ID: mdl-27748374

ABSTRACT

Myeloproliferative neoplasms (MPN), which overproduce blood cells in the bone marrow, have recently been linked with a genetically determined decrease in expression of the MYB transcription factor. Here, we use a mouse MYB knockdown model with an MPN-like phenotype to show how lower levels of MYB lead to stem cell characteristics in myeloid progenitors. The altered progenitor properties feature elevated cytokine responsiveness, especially to interleukin-3, which results from increased receptor expression and increased MAPK activity leading to enhanced phosphorylation of a key regulator of protein synthesis, ribosomal protein S6. MYB acts on MAPK signaling by directly regulating transcription of the gene encoding the negative modulator SPRY2. This mechanistic insight points to pathways that might be targeted therapeutically in MPN.


Subject(s)
Gene Expression Regulation , Interleukin-3/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Myeloid Cells/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Transcription, Genetic , Animals , Biomarkers , Cell Line , Cell Proliferation , Fetal Blood/cytology , Gene Expression , Gene Expression Regulation/drug effects , Humans , Immunophenotyping , Interleukin-3/pharmacology , Models, Molecular , Myeloid Progenitor Cells/drug effects , Phenotype , Receptors, Interleukin-3/metabolism , Signal Transduction/drug effects
19.
J Leukoc Biol ; 101(1): 227-238, 2017 01.
Article in English | MEDLINE | ID: mdl-27443880

ABSTRACT

Cytokines of the GM-CSF family signal via the same receptor subunit (ßc) and, thus, have overlapping effects on cells that express all cytokine-specific α-chains (IL-3Rα, IL-5Rα, GM-CSFRα), such as human basophils, whose rapid effector functions are similarly enhanced by IL-3, IL-5, and GM-CSF. However, previous work has shown that IL-3, but not IL-5 and GM-CSF, supports and induces allergy-associated functions of human basophils at later time points. This includes induction of Th2 cytokine and chemokine secretion, high-affinity IgE receptor-independent leukotriene C4 (LTC4) formation, expression of enzymes (e.g., RALDH2, granzyme B), and kinases (e.g., Pim1). Here, we address the question of why IL-3, but not IL-5 or GM-CSF, is capable of inducing these late responses in human basophils, and we investigate the mechanism that underlies the unique regulatory capacity of IL-3. We find that IL-3, IL-5, and GM-CSF rapidly activate the same canonical signaling cascades in a qualitatively identical manner with comparable strength, but we identify signaling duration as major discriminating factor. IL-5 and GM-CSF rapidly down-regulate surface levels of their receptors within minutes, concomitant with a rapid decay in signaling molecule activation and time-dependent loss of ability of these cytokines to prime basophils for functional responses. By contrast, IL-3 hardly down-regulates the α-chain of its receptor without depleting the common ß-chain, which enables extraordinarily sustained signaling events, predominantly the activation of Stat5. Of interest, acute IL-3 signaling is not sufficient to induce persistent phenotypical and functional changes in human basophils. Induction of these functional late responses depends on continuous IL-3 receptor activation and signaling.


Subject(s)
Basophils/metabolism , Interleukin-3/metabolism , Receptors, Interleukin-3/metabolism , Signal Transduction , Down-Regulation/drug effects , Eosinophils/drug effects , Eosinophils/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-5/metabolism , Kinetics , Ligands , Phenotype , Phosphorylation/drug effects , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
20.
Br J Cancer ; 115(8): 940-948, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27575850

ABSTRACT

BACKGROUND: Only a small proportion of patients respond to anti-VEGF therapy, pressing the need for a reliable biomarker that can identify patients who will benefit. We studied the biological activity of anti-VEGF antibodies in patients' blood during anti-VEGF therapy by using the Ba/F3-VEGFR2 cell line, which is dependent on VEGF for its growth. METHODS: Serum samples from 22 patients with cancer before and during treatment with bevacizumab were tested for their effect on proliferation of Ba/F3-VEGFR2 cells. Vascular endothelial growth factor as well as bevacizumab concentrations in serum samples from these patients were determined by enzyme linked immunosorbent assay (ELISA). RESULTS: The hVEGF-driven cell proliferation was effectively blocked by bevacizumab (IC50 3.7 µg ml-1; 95% CI 1.7-8.3 µg ml-1). Cell proliferation was significantly reduced when patients' serum during treatment with bevacizumab was added (22-103% inhibition compared with pre-treatment). Although bevacizumab levels were not related, on-treatment serum VEGF levels were correlated with Ba/F3-VEGFR2 cell proliferation. CONCLUSIONS: We found that the neutralising effect of anti-VEGF antibody therapy on the biological activity of circulating VEGF can be accurately determined with a Ba/F3-VEGFR2 bioassay. The value of this bioassay to predict clinical benefit of anti-VEGF antibody therapy needs further clinical evaluation in a larger randomised cohort.


Subject(s)
Angiogenesis Inhibitors/blood , B-Lymphocytes/drug effects , Bevacizumab/blood , Biological Assay , Enzyme-Linked Immunosorbent Assay , Neoplasms/blood , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Cell Division , Cell Line , Interleukin-3/pharmacology , Mice , Neoplasms/drug therapy , Receptors, Erythropoietin/genetics , Receptors, Interleukin-3/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Reproducibility of Results , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...