Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
J Leukoc Biol ; 105(6): 1183-1194, 2019 06.
Article in English | MEDLINE | ID: mdl-30942918

ABSTRACT

GM-CSF is important in regulating acute, persistent neutrophilic inflammation in certain settings, including lung injury. Ligand binding induces rapid internalization of the GM-CSF receptor (GM-CSFRα) complex, a process essential for signaling. Whereas GM-CSF controls many aspects of neutrophil biology, regulation of GM-CSFRα expression is poorly understood, particularly the role of GM-CSFRα in ligand clearance and whether signaling is sustained despite major down-regulation of GM-CSFRα surface expression. We established a quantitative assay of GM-CSFRα surface expression and used this, together with selective anti-GM-CSFR antibodies, to define GM-CSFRα kinetics in human neutrophils, and in murine blood and alveolar neutrophils in a lung injury model. Despite rapid sustained ligand-induced GM-CSFRα loss from the neutrophil surface, which persisted even following ligand removal, pro-survival effects of GM-CSF required ongoing ligand-receptor interaction. Neutrophils recruited to the lungs following LPS challenge showed initially high mGM-CSFRα expression, which along with mGM-CSFRß declined over 24 hr; this was associated with a transient increase in bronchoalveolar lavage fluid (BALF) mGM-CSF concentration. Treating mice in an LPS challenge model with CAM-3003, an anti-mGM-CSFRα mAb, inhibited inflammatory cell influx into the lung and maintained the level of BALF mGM-CSF. Consistent with neutrophil consumption of GM-CSF, human neutrophils depleted exogenous GM-CSF, independent of protease activity. These data show that loss of membrane GM-CSFRα following GM-CSF exposure does not preclude sustained GM-CSF/GM-CSFRα signaling and that this receptor plays a key role in ligand clearance. Hence neutrophilic activation via GM-CSFR may play an important role in neutrophilic lung inflammation even in the absence of high GM-CSF levels or GM-CSFRα expression.


Subject(s)
Acute Lung Injury/immunology , Gene Expression Regulation/immunology , Neutrophils/immunology , Pulmonary Alveoli/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Adult , Animals , Cell Line, Tumor , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Disease Models, Animal , Female , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutrophils/pathology , Pulmonary Alveoli/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Time Factors
3.
MAbs ; 8(3): 436-53, 2016.
Article in English | MEDLINE | ID: mdl-26651396

ABSTRACT

The ß common-signaling cytokines interleukin (IL)-3, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-5 stimulate pro-inflammatory activities of haematopoietic cells via a receptor complex incorporating cytokine-specific α and shared ß common (ßc, CD131) receptor. Evidence from animal models and recent clinical trials demonstrate that these cytokines are critical mediators of the pathogenesis of inflammatory airway disease such as asthma. However, no therapeutic agents, other than steroids, that specifically and effectively target inflammation mediated by all 3 of these cytokines exist. We employed phage display technology to identify and optimize a novel, human monoclonal antibody (CSL311) that binds to a unique epitope that is specific to the cytokine-binding site of the human ßc receptor. The binding epitope of CSL311 on the ßc receptor was defined by X-ray crystallography and site-directed mutagenesis. CSL311 has picomolar binding affinity for the human ßc receptor, and at therapeutic concentrations is a highly potent antagonist of the combined activities of IL-3, GM-CSF and IL-5 on primary eosinophil survival in vitro. Importantly, CSL311 inhibited the survival of inflammatory cells present in induced sputum from human allergic asthmatic subjects undergoing allergen bronchoprovocation. Due to its high potency and ability to simultaneously suppress the activity of all 3 ß common cytokines, CSL311 may provide a new strategy for the treatment of chronic inflammatory diseases where the human ßc receptor is central to pathogenesis. The coordinates for the ßc/CSL311 Fab complex structure have been deposited with the RCSB Protein Data Bank (PDB 5DWU).


Subject(s)
Antibodies, Monoclonal, Murine-Derived , Cytokine Receptor Common beta Subunit , Epitopes , Granulocyte-Macrophage Colony-Stimulating Factor , Interleukin-3 , Interleukin-5 , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/immunology , Asthma/drug therapy , Asthma/immunology , Asthma/pathology , Crystallography, X-Ray , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/immunology , Eosinophils/immunology , Eosinophils/pathology , Epitopes/chemistry , Epitopes/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-3/antagonists & inhibitors , Interleukin-3/immunology , Interleukin-5/antagonists & inhibitors , Interleukin-5/immunology , Mice
4.
Immunity ; 43(3): 502-14, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26341401

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) has emerged as a crucial cytokine produced by auto-reactive T helper (Th) cells that initiate tissue inflammation. Multiple cell types can sense GM-CSF, but the identity of the pathogenic GM-CSF-responsive cells is unclear. By using conditional gene targeting, we systematically deleted the GM-CSF receptor (Csf2rb) in specific subpopulations throughout the myeloid lineages. Experimental autoimmune encephalomyelitis (EAE) progressed normally when either classical dendritic cells (cDCs) or neutrophils lacked GM-CSF responsiveness. The development of tissue-invading monocyte-derived dendritic cells (moDCs) was also unperturbed upon Csf2rb deletion. Instead, deletion of Csf2rb in CCR2(+)Ly6C(hi) monocytes phenocopied the EAE resistance seen in complete Csf2rb-deficient mice. High-dimensional analysis of tissue-infiltrating moDCs revealed that GM-CSF initiates a combination of inflammatory mechanisms. These results indicate that GM-CSF signaling controls a pathogenic expression signature in CCR2(+)Ly6C(hi) monocytes and their progeny, which was essential for tissue damage.


Subject(s)
Autoimmunity/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Inflammation/immunology , Monocytes/immunology , Receptors, CCR2/immunology , Signal Transduction/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Antigens, Ly/metabolism , Autoimmunity/genetics , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Cytokine Receptor Common beta Subunit/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice, Knockout , Mice, Transgenic , Monocytes/drug effects , Monocytes/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptome/drug effects , Transcriptome/genetics , Transcriptome/immunology
5.
Cytokine ; 74(2): 247-58, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25982846

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and IL-5 are members of a small family of cytokines that share a beta receptor subunit (ßc). These cytokines regulate the growth, differentiation, migration and effector function activities of many hematopoietic cells in bone marrow, blood and sites of inflammation. Excessive or aberrant signaling can result in chronic inflammatory conditions and myeloid leukemias. The crystal structures of the GM-CSF ternary complex, the IL-5 binary complex and the very recent IL-3 receptor alpha subunit build upon decades of structure-function studies, giving new insights into cytokine-receptor specificity and signal transduction. Selective modulation of receptor function is now a real possibility and the structures of the ßc receptor family are being used to discover novel and disease-specific therapeutics.


Subject(s)
Cytokine Receptor Common beta Subunit , Cytokines , Signal Transduction/immunology , Animals , Cytokine Receptor Common beta Subunit/chemistry , Cytokine Receptor Common beta Subunit/immunology , Cytokines/chemistry , Cytokines/immunology , Humans , Protein Structure, Quaternary , Structure-Activity Relationship
6.
Leuk Res ; 38(11): 1367-73, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25278187

ABSTRACT

Engraftment of primary human acute myeloid leukemia (AML) specimens into the bone marrow (BM) of NOD/SCID mice has been used to study leukemia biology and new treatments for the disease. CSL360 is a chimeric IgG1 monoclonal antibody that recognizes CD123 (IL-3 receptor α-subchain) expressed in the absence of CD131 (ß-subchain), an epitope that is displayed by leukemia stem cells (LSCs). We are studying CSL360 modified with diethylenetriaminepentaacetic acid (DTPA) for complexing 111In and 13-mer nuclear translocation sequence (NLS) peptides to enable nuclear importation in LSCs for Auger electron radioimmunotherapy (RIT) of AML. We demonstrate that microSPECT/CT imaging using 111In-DTPA-NLS-CSL360 revealed engraftment of primary human AML specimens into the BM and spleen of NOD/SCID mice. Our results suggest that microSPECT/CT imaging is a powerful tool which enables non-invasive assessment of the engraftment of AML into NOD/SCID mice and in the current study specifically probes an epitope displayed by the LSC subpopulation. The targeting of 111In-DTPA-NLS-CSL360 to sites of AML engraftment in the NOD/SCID mouse model is encouraging for future RIT studies. Ultimately, SPECT imaging could be applied in AML patients to assess the delivery of 111In-DTPA-NLS-CSL360 to sites of leukemia and be combined with Auger electron RIT using the same agent targeting the LSC population as a "theranostic" pair.


Subject(s)
Bone Marrow/pathology , Immunoconjugates , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/immunology , Spleen/pathology , Tomography, Emission-Computed, Single-Photon , Animals , Cell Line, Tumor , Cytokine Receptor Common beta Subunit/immunology , Humans , Immunoconjugates/pharmacokinetics , Indium Radioisotopes , Interleukin-3 Receptor alpha Subunit/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Multimodal Imaging , Tissue Distribution
7.
PLoS Pathog ; 10(4): e1004053, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24699679

ABSTRACT

Alveolar macrophages (AM) are critical for defense against bacterial and fungal infections. However, a definitive role of AM in viral infections remains unclear. We here report that AM play a key role in survival to influenza and vaccinia virus infection by maintaining lung function and thereby protecting from asphyxiation. Absence of AM in GM-CSF-deficient (Csf2-/-) mice or selective AM depletion in wild-type mice resulted in impaired gas exchange and fatal hypoxia associated with severe morbidity to influenza virus infection, while viral clearance was affected moderately. Virus-induced morbidity was far more severe in Csf2-/- mice lacking AM, as compared to Batf3-deficient mice lacking CD8α+ and CD103+ DCs. Csf2-/- mice showed intact anti-viral CD8+ T cell responses despite slightly impaired CD103+ DC development. Importantly, selective reconstitution of AM development in Csf2rb-/- mice by neonatal transfer of wild-type AM progenitors prevented severe morbidity and mortality, demonstrating that absence of AM alone is responsible for disease severity in mice lacking GM-CSF or its receptor. In addition, CD11c-Cre/Ppargfl/fl mice with a defect in AM but normal adaptive immunity showed increased morbidity and lung failure to influenza virus. Taken together, our results suggest a superior role of AM compared to CD103+ DCs in protection from acute influenza and vaccinia virus infection-induced morbidity and mortality.


Subject(s)
Immunity, Cellular , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/immunology , Respiratory Insufficiency/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Influenza A Virus, H1N1 Subtype , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , PPAR gamma/genetics , PPAR gamma/immunology , Respiratory Insufficiency/genetics , Respiratory Insufficiency/pathology , Respiratory Insufficiency/virology
8.
Immunol Cell Biol ; 90(5): 553-8, 2012 May.
Article in English | MEDLINE | ID: mdl-21844882

ABSTRACT

Granulocyte macrophage-colony stimulating factor (GM-CSF) is a cytokine with the capacity to promote inflammation in a wide variety of infectious and inflammatory diseases. These conditions include allergic airway inflammation, which is driven by T-helper 2 (Th2) cells. Because of the importance of Th2 cells in parasite infections, we have investigated the role of GM-CSF in mice infected with the nematode Nippostrongylus brasiliensis. The effect of primary and secondary infection was investigated in mice lacking functional genes for GM-CSF (CSF2 genes) (ΔGM-CSF mice), and in mice lacking the cytokine receptor common ß chain (Δß mice), the latter being unable to signal in response to GM-CSF and interleukin (IL)-5. ΔGM-CSF mice showed no significant defect in parasite immunity, measured by larval numbers in the lungs, worm numbers in the intestine or egg numbers in the faeces, in either primary or secondary infection. By contrast, the Δß mice showed increased parasite burden, with higher numbers of lung larvae after secondary infection and higher numbers of intestinal worms and faecal eggs after both primary and secondary infection. Unexpectedly, there were increased numbers of circulating eosinophils in the ΔGM-CSF mice, associated with significantly reduced larval numbers in the lungs. These results indicate that GM-CSF is redundant in protection against N. brasiliensis infection, and that the increased susceptibility of Δß mice to infection is likely to be attributed to the lack of IL-5 signalling in these mice. The results suggest that clinical use of agents that neutralise GM-CSF may not be associated with increased risk of parasite infection.


Subject(s)
Cytokine Receptor Common beta Subunit/metabolism , Eosinophils/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Nippostrongylus/immunology , Strongylida Infections/immunology , Strongylida Infections/prevention & control , Animals , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoglobulin E/blood , Intestines/immunology , Intestines/parasitology , Larva , Lung/immunology , Lung/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasite Load , Th2 Cells/immunology
9.
J Cell Physiol ; 226(12): 3330-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21321940

ABSTRACT

Erythropoietin (EPO), the key hormone for erythropoiesis, also increases nitric oxide (NO) bioavailability in endothelial cells (ECs), yet the definitive mechanisms are not fully understood. Increasing evidence has demonstrated that ß common receptor (ßCR) plays a crucial role in EPO-mediated non-hematopoietic effects. We investigated the role of ßCR in EPO-induced endothelial NO synthase (eNOS) activation in bovine aortic ECs (BAECs) and the molecular mechanisms involved. Results of confocal microscopy and immunoprecipitation analyses revealed that ßCR was colocalized and interacted with EPO receptor (EPOR) in ECs. Inhibition of ßCR or EPOR by neutralizing antibodies or small interfering RNA abolished the EPO-induced NO production. Additionally, blockage of ßCR abrogated the EPO-induced increase in the phosphorylation of eNOS, Akt, Src, or Janus kinase 2 (JAK2). Immunoprecipitation analysis revealed that treatment with EPO increased the interaction between ßCR and eNOS, which was suppressed by inhibition of Src, JAK2, or Akt signaling with specific pharmacological inhibitors. Furthermore, EPO-induced EC proliferation, migration, and tube formation were blocked by pretreatment with ßCR antibody and Src, JAK2, or PI3K/Akt inhibitors. Moreover, in vivo experiments showed that EPO increased the level of phosphorylated eNOS, Src, JAK2, and Akt, as well as ßCR-eNOS association in aortas and promoted the angiogenesis in Matrigel plug, which was diminished by ßCR or EPOR neutralizing antibodies. Our findings suggest that ßCR may play an integrative role in the EPO signaling-mediated activation of eNOS in ECs.


Subject(s)
Cytokine Receptor Common beta Subunit/metabolism , Endothelial Cells/enzymology , Erythropoietin/metabolism , Nitric Oxide Synthase Type III/metabolism , Receptors, Interleukin-3/metabolism , Signal Transduction , Animals , Antibodies, Neutralizing/pharmacology , Cattle , Cell Movement , Cell Proliferation , Cells, Cultured , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/immunology , Endothelial Cells/drug effects , Enzyme Activation , Erythropoietin/genetics , Humans , Immunoprecipitation , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Neovascularization, Physiologic , Nitric Oxide/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/immunology , Receptors, Erythropoietin/metabolism , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/immunology , Recombinant Proteins , Signal Transduction/drug effects , Time Factors , Transfection , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL