Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
Cell Mol Immunol ; 21(1): 6-18, 2024 01.
Article in English | MEDLINE | ID: mdl-38114747

ABSTRACT

Emergency granulopoiesis and neutrophil mobilization that can be triggered by granulocyte colony-stimulating factor (G-CSF) through its receptor G-CSFR are essential for antibacterial innate defense. However, the epigenetic modifiers crucial for intrinsically regulating G-CSFR expression and the antibacterial response of neutrophils remain largely unclear. N6-methyladenosine (m6A) RNA modification and the related demethylase alkB homolog 5 (ALKBH5) are key epigenetic regulators of immunity and inflammation, but their roles in neutrophil production and mobilization are still unknown. We used cecal ligation and puncture (CLP)-induced polymicrobial sepsis to model systemic bacterial infection, and we report that ALKBH5 is required for emergency granulopoiesis and neutrophil mobilization. ALKBH5 depletion significantly impaired the production of immature neutrophils in the bone marrow of septic mice. In addition, Alkbh5-deficient septic mice exhibited higher retention of mature neutrophils in the bone marrow and defective neutrophil release into the circulation, which led to fewer neutrophils at the infection site than in their wild-type littermates. During bacterial infection, ALKBH5 imprinted production- and mobilization-promoting transcriptome signatures in both mouse and human neutrophils. Mechanistically, ALKBH5 erased m6A methylation on the CSF3R mRNA to increase the mRNA stability and protein expression of G-CSFR, consequently upregulating cell surface G-CSFR expression and downstream STAT3 signaling in neutrophils. The RIP-qPCR results confirmed the direct binding of ALKBH5 to the CSF3R mRNA, and the binding strength declined upon bacterial infection, accounting for the decrease in G-CSFR expression on bacteria-infected neutrophils. Considering these results collectively, we define a new role of ALKBH5 in intrinsically driving neutrophil production and mobilization through m6A demethylation-dependent posttranscriptional regulation, indicating that m6A RNA modification in neutrophils is a potential target for treating bacterial infections and neutropenia.


Subject(s)
Bacterial Infections , Sepsis , Animals , Humans , Mice , AlkB Homolog 5, RNA Demethylase/metabolism , Anti-Bacterial Agents , Neutrophils , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , RNA/metabolism , RNA, Messenger/metabolism
2.
J Immunol ; 208(5): 1066-1075, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35140132

ABSTRACT

BATF3-deficient mice that lack CD8+ dendritic cells (DCs) showed an exacerbation of chronic graft-versus-host disease (cGVHD), including T follicular helper (Tfh) cell and autoantibody responses, whereas mice carrying the Sle2c2 lupus-suppressive locus with a mutation in the G-CSFR showed an expansion of CD8+ DCs and a poor mobilization of plasmacytoid DCs (pDCs) and responded poorly to cGVHD induction. Here, we investigated the contribution of CD8+ DCs and pDCs to the humoral response to protein immunization, where CD8neg DCs are thought to represent the major inducers. Both BATF3-/- and Sle2c2 mice had reduced humoral and germinal center (GC) responses compared with C57BL/6 (B6) controls. We showed that B6-derived CD4+ DCs are the major early producers of IL-6, followed by CD4-CD8- DCs. Surprisingly, IL-6 production and CD80 expression also increased in CD8+ DCs after immunization, and B6-derived CD8+ DCs rescued Ag-specific adaptive responses in BATF3-/- mice. In addition, inflammatory pDCs (ipDCs) produced more IL-6 than all conventional DCs combined. Interestingly, G-CSFR is highly expressed on pDCs. G-CSF expanded pDC and CD8+ DC numbers and IL-6 production by ipDCs and CD4+ DCs, and it improved the quality of Ab response, increasing the localization of Ag-specific T cells to the GC. Finally, G-CSF activated STAT3 in early G-CSFR+ common lymphoid progenitors of cDCs/pDCs but not in mature cells. In conclusion, we showed a multilayered role of DC subsets in priming Tfh cells in protein immunization, and we unveiled the importance of G-CSFR signaling in the development and function pDCs.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Lymphoid Progenitor Cells/cytology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , T Follicular Helper Cells/immunology , Adoptive Transfer , Animals , Autoantibodies/immunology , B7-1 Antigen/biosynthesis , Basic-Leucine Zipper Transcription Factors/genetics , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cell Differentiation/immunology , Dendritic Cells/transplantation , Female , Granulocyte Colony-Stimulating Factor/metabolism , Interleukin-6/biosynthesis , Lymphocyte Activation/immunology , Lymphoid Progenitor Cells/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology
3.
Front Immunol ; 13: 1095453, 2022.
Article in English | MEDLINE | ID: mdl-36703974

ABSTRACT

Introduction: The granulocyte colony-stimulating factor receptor (G-CSFR), encoded by the CSF3R gene, is involved in the production and function of neutrophilic granulocytes. Somatic mutations in CSF3R leading to truncated G-CSFR forms are observed in acute myeloid leukemia (AML), particularly those subsequent to severe chronic neutropenia (SCN), as well as in a subset of patients with other leukemias. Methods: This investigation introduced equivalent mutations into the zebrafish csf3r gene via genome editing and used a range of molecular and cellular techniques to understand the impact of these mutations on immune cells across the lifespan. Results: Zebrafish harboring truncated G-CSFRs showed significantly enhanced neutrophil production throughout successive waves of embryonic hematopoiesis and a neutrophil maturation defect in adults, with the mutations acting in a partially dominant manner. Discussion: This study has elucidated new insights into the impact of G-CSFR truncations throughout the life-course and created a bone fide zebrafish model for further investigation.


Subject(s)
Hematopoiesis , Receptors, Granulocyte Colony-Stimulating Factor , Animals , Hematopoiesis/genetics , Leukemia, Myeloid, Acute/genetics , Leukopoiesis/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Zebrafish
4.
Semin Immunol ; 54: 101515, 2021 04.
Article in English | MEDLINE | ID: mdl-34772606

ABSTRACT

A considerable amount of continuous proliferation and differentiation is required to produce daily a billion new neutrophils in an adult human. Of the few cytokines and factors known to control neutrophil production, G-CSF is the guardian of granulopoiesis. G-CSF/CSF3R signaling involves the recruitment of non-receptor protein tyrosine kinases and their dependent signaling pathways of serine/threonine kinases, tyrosine phosphatases, and lipid second messengers. These pathways converge to activate the families of STAT and C/EBP transcription factors. CSF3R mutations are associated with human disorders of neutrophil production, including severe congenital neutropenia, neutrophilia, and myeloid malignancies. More than three decades after their identification, cloning, and characterization of G-CSF and G-CSF receptor, fundamental questions remain about their physiology.


Subject(s)
Granulocyte Colony-Stimulating Factor , Neutropenia , Adult , Congenital Bone Marrow Failure Syndromes , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis , Humans , Neutropenia/congenital , Neutropenia/genetics , Neutropenia/pathology , Neutrophils/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism
5.
Biotechnol J ; 15(2): e1900052, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31469473

ABSTRACT

Producing a sufficient number of cardiomyocytes from pluripotent stem cells has been of great demand for cardiac regeneration therapy. However, it remains challenging to efficiently differentiate cardiomyocytes with low costs. Reportedly, granulocyte colony-stimulating factor (G-CSF) receptor (GCSFR) signaling activates signal transducers and activators of transcription (STAT) signaling and enhances cardiac differentiation from embryonic stem cells or induced pluripotent stem cells (iPSCs). To economically and efficiently produce cardiomyocytes from iPSCs through GCSFR/STAT axis activation, we constructed antibody/receptor chimeras that can respond to an inexpensive small molecule. Single-chain Fv of anti-fluorescein (FL) antibody was ligated to transmembrane/cytoplasmic domains of GCSFRs, enabling transduction of GCSFR signaling in response to FL-conjugated bovine serum albumin (BSA-FL) as an alternative ligand. Mouse iPSC lines constitutively expressing these chimeric receptors exhibited increased BSA-FL-induced STAT3 phosphorylation in a dose-dependent manner, which was abolished by an inhibitor of Janus tyrosine kinase (JAK). In addition, BSA-FL stimulation also increased the incidence of beating embryoid bodies and upregulated cardiac-specific gene expressions after differentiation in these iPSC lines. Therefore, the chimeric GCSFRs activated endogenous GCSFR signaling at least via the JAK/STAT3 pathway, thereby enhancing cardiac differentiation from iPSCs. This approach, as an economical strategy, could contribute to stem cell-based cardiac regeneration therapy.


Subject(s)
Janus Kinase 1/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Differentiation , Female , Induced Pluripotent Stem Cells/physiology , Janus Kinase 1/genetics , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/physiology , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Fusion Proteins , STAT3 Transcription Factor/genetics
6.
Sci Rep ; 9(1): 17732, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31776393

ABSTRACT

Excessive neutrophilic inflammation can contribute to the pathogenesis of pneumonia. Whilst anti-inflammatory therapies such as corticosteroids are used to treat excessive inflammation, they do not selectively target neutrophils and may compromise antimicrobial or antiviral defences. In this study, neutrophil trafficking was targeted with a granulocyte-colony stimulating factor receptor monoclonal antibody (G-CSFR mAb) during Streptococcus pneumoniae (serotype 19F) or influenza A virus (IAV, strain HKx31) lung infection in mice. Firstly, we demonstrated that neutrophils are indispensable for the clearance of S. pneumoniae from the airways using an anti-Ly6G monoclonal antibody (1A8 mAb), as the complete inhibition of neutrophil recruitment markedly compromised bacterial clearance. Secondly, we demonstrated that G-CSF transcript lung levels were significantly increased during pneumococcal infection. Prophylactic or therapeutic administration of G-CSFR mAb significantly reduced blood and airway neutrophil numbers by 30-60% without affecting bacterial clearance. Total protein levels in the bronchoalveolar lavage (BAL) fluid (marker for oedema) was also significantly reduced. G-CSF transcript levels were also increased during IAV lung infection. G-CSFR mAb treatment significantly reduced neutrophil trafficking into BAL compartment by 60% and reduced blood neutrophil numbers to control levels in IAV-infected mice. Peak lung viral levels at day 3 were not altered by G-CSFR therapy, however there was a significant reduction in the detection of IAV in the lungs at the day 7 post-infection phase. In summary, G-CSFR signalling contributes to neutrophil trafficking in response to two common respiratory pathogens. Blocking G-CSFR reduced neutrophil trafficking and oedema without compromising clearance of two pathogens that can cause pneumonia.


Subject(s)
Neutrophil Infiltration , Neutrophils/immunology , Orthomyxoviridae Infections/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Granulocyte Colony-Stimulating Factor/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Female , Lung/metabolism , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism
7.
Commun Biol ; 2: 45, 2019.
Article in English | MEDLINE | ID: mdl-30729183

ABSTRACT

During bacterial infection, granulocyte colony-stimulating factor (G-CSF) is produced and accelerates neutrophil production from their progenitors. This process, termed granulopoiesis, strengthens host defense, but Clostridium perfringens α-toxin impairs granulopoiesis via an unknown mechanism. Here, we tested whether G-CSF accounts for the α-toxin-mediated impairment of granulopoiesis. We find that α-toxin dramatically accelerates G-CSF production from endothelial cells in response to Toll-like receptor 2 (TLR2) agonists through activation of the c-Jun N-terminal kinase (JNK) signaling pathway. Meanwhile, α-toxin inhibits G-CSF-mediated cell proliferation of Ly-6G+ neutrophils by inducing degradation of G-CSF receptor (G-CSFR). During sepsis, administration of α-toxin promotes lethality and tissue injury accompanied by accelerated production of inflammatory cytokines in a TLR4-dependent manner. Together, our results illustrate that α-toxin disturbs G-CSF-mediated granulopoiesis by reducing the expression of G-CSFR on neutrophils while augmenting septic shock due to excess inflammatory cytokine release, which provides a new mechanism to explain how pathogenic bacteria modulate the host immune system.


Subject(s)
Bacterial Toxins/toxicity , Calcium-Binding Proteins/toxicity , Clostridium perfringens/pathogenicity , Gas Gangrene/genetics , Granulocyte Colony-Stimulating Factor/genetics , Lipopolysaccharides/toxicity , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Shock, Septic/genetics , Type C Phospholipases/toxicity , Animals , Clostridium perfringens/genetics , Clostridium perfringens/immunology , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Female , Gas Gangrene/immunology , Gas Gangrene/microbiology , Gas Gangrene/mortality , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/immunology , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoiesis/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/microbiology , Receptors, Granulocyte Colony-Stimulating Factor/immunology , Shock, Septic/immunology , Shock, Septic/microbiology , Shock, Septic/mortality , Signal Transduction , Survival Analysis , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
8.
Sci Rep ; 9(1): 747, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30679689

ABSTRACT

Ceramides are sphingolipids with defined acyl chain lengths, which are produced by corresponding ceramide synthases (CerS1-6). In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), the ablation of CerS2 suppresses EAE-pathology by reducing neutrophil migration into the central nervous system. This migration is induced by granulocyte-colony stimulating factor (G-CSF) signaling. G-CSF signaling leads to a signal cascade including the phosphorylation of Lyn kinase and STAT3. This in turn regulates expression of the neutrophil surface receptor chemokine receptor 2 (CXCR2) and causes translocation of the receptor into detergent-resistant membranes (DRMs). In this study we investigated the role of ceramides in G-CSF signaling. We found, that G-CSF treatment of wild type bone marrow cells (BMCs) leads to translocation of G-CSF-receptor (G-CSF-R) into DRMs. G-CSF also induces downregulation of ceramides in WT and CerS2 null BMCs, as well as upregulation of very long chain lactosylceramides. However, in CerS2 null BMCs, G-CSF failed to induce translocation of G-CSF-R into DRMs, leading to reduced phosphorylation of Lyn and reduced CXCR2 expression. Interestingly, G-CSF signaling in CerS6 null BMCs was not affected. In conclusion, very long chain ceramides are important for G-CSF signaling and translocation of G-CSF-R into DRMs.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Granulocyte Colony-Stimulating Factor/genetics , Multiple Sclerosis/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Sphingosine N-Acyltransferase/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Movement/drug effects , Detergents/pharmacology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/drug effects , Humans , Lactosylceramides/metabolism , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neutrophils/drug effects , Protein Transport/drug effects , STAT3 Transcription Factor/genetics , src-Family Kinases/genetics
9.
Leukemia ; 33(1): 75-87, 2019 01.
Article in English | MEDLINE | ID: mdl-29977015

ABSTRACT

Granulocyte-colony stimulating factor receptor (G-CSFR) controls myeloid progenitor proliferation and differentiation to neutrophils. Mutations in CSF3R (encoding G-CSFR) have been reported in patients with chronic neutrophilic leukemia (CNL) and acute myeloid leukemia (AML); however, despite years of research, the malignant downstream signaling of the mutated G-CSFRs is not well understood. Here, we used a quantitative phospho-tyrosine analysis to generate a comprehensive signaling map of G-CSF induced tyrosine phosphorylation in the normal versus mutated (proximal: T618I and truncated: Q741x) G-CSFRs. Unbiased clustering and kinase enrichment analysis identified rapid induction of phospho-proteins associated with endocytosis by the wild type G-CSFR only; while G-CSFR mutants showed abnormal kinetics of canonical Stat3, Stat5, and Mapk phosphorylation, and aberrant activation of Bruton's Tyrosine Kinase (Btk). Mutant-G-CSFR-expressing cells displayed enhanced sensitivity (3-5-fold lower IC50) for ibrutinib-based chemical inhibition of Btk. Primary murine progenitor cells from G-CSFR-Q741x knock-in mice validated activation of Btk by the mutant receptor and retrovirally transduced human CD34+ umbilical cord blood cells expressing mutant receptors displayed enhanced sensitivity to Ibrutinib. A significantly lower clonogenic potential was displayed by both murine and human primary cells expressing mutated receptors upon ibrutinib treatment. Finally, a dramatic synergy was observed between ibrutinib and ruxolinitib at lower dose of the individual drug. Altogether, these data demonstrate the strength of unsupervised proteomics analyses in dissecting oncogenic pathways, and suggest repositioning Ibrutinib for therapy of myeloid leukemia bearing CSF3R mutations. Phospho-tyrosine data are available via ProteomeXchange with identifier PXD009662.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Mutation , Phosphoproteins/metabolism , Precursor Cells, B-Lymphoid/metabolism , Proteome/analysis , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Humans , Leukemia, Myeloid, Acute , Leukemia, Neutrophilic, Chronic , Mice , Phosphorylation , Piperidines , Precursor Cells, B-Lymphoid/pathology , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism
10.
Exp Oncol ; 40(4): 328-331, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30593760

ABSTRACT

BACKGROUND: Chronic myeloid leukemia (CML) is a clonal hematopoietic stem cell disorder associated with the activity of BCR-ABL fusion oncogene. Tyrosine kinase inhibitors are the current treatment of CML, but secondary mutations finally contribute to therapy resistance and blast crisis of the disease. The search for the novel compounds for the effective control of CML is now in the spotlight. The progression of CML to blast crisis is correlated with down-modulation of C/EBP alpha. Therefore, C/EBP alpha may be considered as a putative target in differentiation therapies in myeloid leukemias. The aim of the study was to assess the potential of vitamin E as the possible inducer of C/EBP alpha expression in BCR-ABL-positive CML K562 cells. MATERIALS AND METHODS: RNA extracted from K562 cells cultured with valproic acid or vitamin E was converted to cDNA, RT-PCR reactions were carried out using HotStarTaq DNA polymerase with primers for C/EBP alpha and granulocyte colony-stimulating factor receptor (G-CSFR). RESULTS: We have not found detectable expression of C/EBP alpha in K562 cells. Upon 48-h culture with vitamin E at a dose of 100 µM, K562 cells expressed both C/EBP alpha and G-CSFR. CONCLUSION: Vitamin E restored the expression of C/EBP alpha mRNA in chronic myelogenous leukemia K562 cells. In this setting, G-CSFR expression in vitamin E treated K562 cells seems to suggest the activation to granulocytic differentiation. It should be further elucidated whether such effects of vitamin E on C/EBP alpha transcription factor are direct or mediated indirectly due to antioxidant properties of vitamin E.


Subject(s)
Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Vitamin E/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/biosynthesis , Transcriptional Activation/drug effects , Valproic Acid/pharmacology
11.
Clin Sci (Lond) ; 132(17): 1963-1976, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30219773

ABSTRACT

Ceramide synthases (CerS) synthesize chain length specific ceramides (Cer), which mediate cellular processes in a chain length-dependent manner. In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), we observed that the genetic deletion of CerS2 suppresses EAE pathology by interaction with granulocyte-colony stimulating factor (G-CSF) signaling and CXC motif chemokine receptor 2 (CXCR2) expression, leading to impaired neutrophil migration. In the present study, we investigated the importance of Cers and their synthesizing/metabolizing enzymes in MS. For this purpose, a longitudinal study with 72 MS patients and 25 healthy volunteers was performed. Blood samples were collected from healthy controls and MS patients over 1- or 3-year periods, respectively. Immune cells were counted using flow cytometry, ceramide levels were determined using liquid chromatography-tandem mass spectrometry, and mRNA expression was analyzed using quantitative PCR. In white blood cells, C16-LacCer and C24-Cer were down-regulated in MS patients in comparison with healthy controls. In plasma, C16-Cer, C24:1-Cer, C16-GluCer, and C24:1-GluCer were up-regulated and C16-LacCer was down-regulated in MS patients in comparison with healthy controls. Blood samples from MS patients were characterized by an increased B-cell number. However, there was no correlation between B-cell number and Cer levels. mRNA expression of Cer metabolizing enzymes and G-CSF signaling enzymes was significantly increased in MS patients. Interestingly, G-CSF receptor (G-CSFR) and CXCR2 mRNA expression correlated with CerS2 and UDP-glucose Cer glucosyltransferase (UGCG) mRNA expression. In conclusion, our results indicate that Cer metabolism is linked to G-CSF signaling in MS.


Subject(s)
Ceramides/blood , Membrane Proteins/metabolism , Multiple Sclerosis/blood , Multiple Sclerosis/metabolism , Sphingosine N-Acyltransferase/metabolism , Tumor Suppressor Proteins/metabolism , B-Lymphocytes/metabolism , Ceramides/chemistry , Ceramides/metabolism , Gene Expression/drug effects , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Leukocyte Count , Leukocytes/metabolism , Longitudinal Studies , Membrane Proteins/genetics , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Multiple Sclerosis/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction , Sphingosine N-Acyltransferase/genetics , Tumor Suppressor Proteins/genetics
12.
Front Immunol ; 9: 1053, 2018.
Article in English | MEDLINE | ID: mdl-29868014

ABSTRACT

The genetic analysis of the lupus-prone NZM2410 mouse has identified a suppressor locus, Sle2c2, which confers resistance to spontaneous lupus in combination with NZM2410 susceptibility loci, or in the chronic graft-versus-host disease (cGVHD) induced model of lupus in the B6.Sle2c2 congenic strain. The candidate gene for Sle2c2, the Csf3r gene encoding the granulocyte colony-stimulating factor receptor (G-CSF-R/CD114), was validated when cGVHD was restored in B6.Sle2c2 mice after treatment with G-CSF. The goal of the project reported herein was to investigate the myeloid cells that confer resistance to cGVHD and to ascertain if the mechanism behind their suppression involves the G-CSF pathway. We showed that despite expressing the highest levels of G-CSF-R, neutrophils play only a modest role in the autoimmune activation induced by cGVHD. We also found reduced expression levels of G-CSF-R on the surface of dendritic cells (DCs) and a differential distribution of DC subsets in response to cGVHD in B6.Sle2c2 versus B6 mice. The CD8α+ DC subset, known for its tolerogenic phenotype, was expanded upon induction of cGVHD in B6.Sle2c2 mice. In addition, the deficiency of CD8α+ DC subset enhanced the severity of cGVHD in B6.Batf3-/- and B6.Sle2c2 mice, confirming their role in suppression of cGVHD. B6.Sle2c2DCs presented lowered activation and antigen presentation abilities and expressed lower levels of genes associated with DC activation and maturation. Exposure to exogenous G-CSF reversed the majority of these phenotypes, suggesting that tolerogenic DCs maintained through a defective G-CSF-R pathway mediated the resistance to cGVHD in B6.Sle2c2 mice.


Subject(s)
Graft vs Host Disease/immunology , Lupus Erythematosus, Systemic/immunology , Myeloid Cells/immunology , Polymorphism, Genetic , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Animals , Dendritic Cells/immunology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Receptors, Granulocyte Colony-Stimulating Factor/therapeutic use
13.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1545-1553, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28578910

ABSTRACT

Granulocyte colony-stimulating factor receptor (G-CSFR) plays a crucial role in regulating myeloid cell survival, proliferation, and neutrophilic granulocyte precursor cells maturation. Previously, we demonstrated that Fbw7α negatively regulates G-CSFR and its downstream signaling through ubiquitin-proteasome mediated degradation. However, whether additional ubiquitin ligases for G-CSFR exist is not known. Identifying multiple E3 ubiquitin ligases for G-CSFR shall improve our understanding of activation and subsequent attenuation of G-CSFR signaling required for differentiation and proliferation. Here, for the first time we demonstrate that E6 associated protein (E6AP), an E3 ubiquitin ligase physically associates with G-CSFR and targets it for ubiquitin-mediated proteasome degradation and thereby attenuates its functions. We further show that E6AP promoted G-CSFR degradation leads to reduced phosphorylation of signal transducer and activator of transcription 3 (STAT3) which is required for G-CSF dependent granulocytic differentiation. More importantly, our finding shows that E6AP also targets mutant form of G-SCFR (G-CSFR-T718), frequently observed in severe congenital neutropenia (SCN) patients that very often culminate to AML, however, at a quite slower rate than wild type G-CSFR. In addition, our data showed that knockdown of E6AP restores G-CSFR and its signaling thereby promoting granulocytic differentiation. Collectively, our data demonstrates that E6AP facilitates ubiquitination and subsequent degradation of G-CSFR leading to attenuation of its downstream signaling and inhibition of granulocytic differentiation.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Ubiquitin-Protein Ligases/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Gene Knockdown Techniques , Granulocytes/metabolism , Granulocytes/pathology , Humans , Myeloid Cells/metabolism , Myeloid Cells/pathology , Proteasome Endopeptidase Complex/genetics , Proteolysis , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
14.
J Immunol ; 198(9): 3565-3575, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28320832

ABSTRACT

G-CSF or CSF-3, originally defined as a regulator of granulocyte lineage development via its cell surface receptor (G-CSFR), can play a role in inflammation, and hence in many pathologies, due to its effects on mature lineage populations. Given this, and because pain is an extremely important arthritis symptom, the efficacy of an anti-G-CSFR mAb for arthritic pain and disease was compared with that of a neutrophil-depleting mAb, anti-Ly6G, in both adaptive and innate immune-mediated murine models. Pain and disease were ameliorated in Ag-induced arthritis, zymosan-induced arthritis, and methylated BSA/IL-1 arthritis by both prophylactic and therapeutic anti-G-CSFR mAb treatment, whereas only prophylactic anti-Ly6G mAb treatment was effective. Efficacy for pain and disease correlated with reduced joint neutrophil numbers and, importantly, benefits were noted without necessarily the concomitant reduction in circulating neutrophils. Anti-G-CSFR mAb also suppressed zymosan-induced inflammatory pain. A new G-CSF-driven (methylated BSA/G-CSF) arthritis model was established enabling us to demonstrate that pain was blocked by a cyclooxygenase-2 inhibitor, suggesting an indirect effect on neurons. Correspondingly, dorsal root ganglion neurons cultured in G-CSF failed to respond to G-CSF in vitro, and Csf3r gene expression could not be detected in dorsal root ganglion neurons by single-cell RT-PCR. These data suggest that G-CSFR/G-CSF targeting may be a safe therapeutic strategy for arthritis and other inflammatory conditions, particularly those in which pain is important, as well as for inflammatory pain per se.


Subject(s)
Antibodies, Blocking/therapeutic use , Arthritis, Experimental/therapy , Arthritis, Rheumatoid/therapy , Immunotherapy/methods , Neurons/drug effects , Neutrophils/immunology , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Animals , Antigens, Ly/immunology , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Cells, Cultured , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/metabolism , Humans , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Neurons/physiology , Neutrophils/drug effects , Neutrophils/pathology , Pain Management , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/immunology
15.
Blood ; 129(6): 667-679, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28028029

ABSTRACT

The genetic landscape of classical myeloproliferative neoplasm (MPN) is in large part elucidated. The MPN-restricted driver mutations, including those in JAK2, calreticulin (CALR), and myeloproliferative leukemia virus (MPL), abnormally activate the cytokine receptor/JAK2 pathway and their downstream effectors, more particularly the STATs. The most frequent mutation, JAK2V617F, activates the 3 main myeloid cytokine receptors (erythropoietin receptor, granulocyte colony-stimulating factor receptor, and MPL) whereas CALR or MPL mutants are restricted to MPL activation. This explains why JAK2V617F is associated with polycythemia vera, essential thrombocythemia (ET), and primary myelofibrosis (PMF) whereas CALR and MPL mutants are found in ET and PMF. Other mutations in genes involved in epigenetic regulation, splicing, and signaling cooperate with the 3 MPN drivers and play a key role in the PMF pathogenesis. Mutations in epigenetic regulators TET2 and DNMT3A are involved in disease initiation and may precede the acquisition of JAK2V617F. Other mutations in epigenetic regulators such as EZH2 and ASXL1 also play a role in disease initiation and disease progression. Mutations in the splicing machinery are predominantly found in PMF and are implicated in the development of anemia or pancytopenia. Both heterogeneity of classical MPNs and prognosis are determined by a specific genomic landscape, that is, type of MPN driver mutations, association with other mutations, and their order of acquisition. However, factors other than somatic mutations play an important role in disease initiation as well as disease progression such as germ line predisposition, inflammation, and aging. Delineation of these environmental factors will be important to better understand the precise pathogenesis of MPN.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Mutation , Polycythemia Vera/genetics , Primary Myelofibrosis/genetics , Thrombocythemia, Essential/genetics , Calreticulin/genetics , Calreticulin/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Disease Progression , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Polycythemia Vera/metabolism , Polycythemia Vera/physiopathology , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/physiopathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Thrombocythemia, Essential/metabolism , Thrombocythemia, Essential/physiopathology
16.
Exp Hematol ; 46: 9-20, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27789332

ABSTRACT

Granulocyte colony-stimulating factor is a hematopoietic cytokine that stimulates neutrophil production and hematopoietic stem cell mobilization by initiating the dimerization of homodimeric granulocyte colony-stimulating factor receptor. Different mutations of CSF3R have been linked to a unique spectrum of myeloid disorders and related malignancies. Myeloid disorders caused by the CSF3R mutations include severe congenital neutropenia, chronic neutrophilic leukemia, and atypical chronic myeloid leukemia. In this review, we provide an analysis of granulocyte colony-stimulating factor receptor, various mutations, and their roles in the severe congenital neutropenia, chronic neutrophilic leukemia, and malignant transformation, as well as the clinical implications and some perspective on approaches that could expand our knowledge with respect to the normal signaling mechanisms and those associated with mutations in the receptor.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Leukemia, Neutrophilic, Chronic/etiology , Leukemia, Neutrophilic, Chronic/metabolism , Neutropenia/congenital , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Signal Transduction , Animals , Cell Transformation, Neoplastic/genetics , Congenital Bone Marrow Failure Syndromes , Genetic Predisposition to Disease , Humans , Janus Kinases/metabolism , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Signaling System , Mutation , Neutropenia/etiology , Neutropenia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Protein Interaction Domains and Motifs/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/chemistry , Receptors, Granulocyte Colony-Stimulating Factor/genetics , STAT Transcription Factors/metabolism
17.
J Immunol ; 197(11): 4392-4402, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27807194

ABSTRACT

G-CSF is a hemopoietic growth factor that has a role in steady state granulopoiesis, as well as in mature neutrophil activation and function. G-CSF- and G-CSF receptor-deficient mice are profoundly protected in several models of rheumatoid arthritis, and Ab blockade of G-CSF also protects against disease. To further investigate the actions of blocking G-CSF/G-CSF receptor signaling in inflammatory disease, and as a prelude to human studies of the same approach, we developed a neutralizing mAb to the murine G-CSF receptor, which potently antagonizes binding of murine G-CSF and thereby inhibits STAT3 phosphorylation and G-CSF receptor signaling. Anti-G-CSF receptor rapidly halted the progression of established disease in collagen Ab-induced arthritis in mice. Neutrophil accumulation in joints was inhibited, without rendering animals neutropenic, suggesting an effect of G-CSF receptor blockade on neutrophil homing to inflammatory sites. Consistent with this, neutrophils in the blood and arthritic joints of anti-G-CSF receptor-treated mice showed alterations in cell adhesion receptors, with reduced CXCR2 and increased CD62L expression. Furthermore, blocking neutrophil trafficking with anti-G-CSF receptor suppressed local production of proinflammatory cytokines (IL-1ß, IL-6) and chemokines (KC, MCP-1) known to drive tissue damage. Differential gene expression analysis of joint neutrophils showed a switch away from an inflammatory phenotype following anti-G-CSF receptor therapy in collagen Ab-induced arthritis. Importantly, G-CSF receptor blockade did not adversely affect viral clearance during influenza infection in mice. To our knowledge, we describe for the first time the effect of G-CSF receptor blockade in a therapeutic model of inflammatory joint disease and provide support for pursuing this therapeutic approach in treating neutrophil-associated inflammatory diseases.


Subject(s)
Antibodies, Neutralizing/pharmacology , Arthritis, Experimental/drug therapy , Gene Expression Regulation/drug effects , Neutrophil Infiltration/drug effects , Neutrophils/immunology , Receptors, Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation/immunology , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/immunology , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Joints/immunology , Joints/pathology , Male , Mice , Mice, Knockout , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/pathology , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/immunology
18.
Bull Math Biol ; 78(12): 2304-2357, 2016 12.
Article in English | MEDLINE | ID: mdl-27324993

ABSTRACT

We develop a physiological model of granulopoiesis which includes explicit modelling of the kinetics of the cytokine granulocyte colony-stimulating factor (G-CSF) incorporating both the freely circulating concentration and the concentration of the cytokine bound to mature neutrophils. G-CSF concentrations are used to directly regulate neutrophil production, with the rate of differentiation of stem cells to neutrophil precursors, the effective proliferation rate in mitosis, the maturation time, and the release rate from the mature marrow reservoir into circulation all dependent on the level of G-CSF in the system. The dependence of the maturation time on the cytokine concentration introduces a state-dependent delay into our differential equation model, and we show how this is derived from an age-structured partial differential equation model of the mitosis and maturation and also detail the derivation of the rest of our model. The model and its estimated parameters are shown to successfully predict the neutrophil and G-CSF responses to a variety of treatment scenarios, including the combined administration of chemotherapy and exogenous G-CSF. This concomitant treatment was reproduced without any additional fitting to characterize drug-drug interactions.


Subject(s)
Granulocyte Colony-Stimulating Factor/physiology , Hematopoiesis/physiology , Neutrophils/cytology , Neutrophils/physiology , Animals , Feedback, Physiological , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/pharmacokinetics , Hematopoiesis/drug effects , Humans , Mathematical Concepts , Mice , Mice, Knockout , Models, Biological , Neutrophils/drug effects , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/physiology
19.
Cytokine ; 78: 69-78, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26687628

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is a pleiotropic cytokine best known for its role in promoting the generation and function of neutrophils. G-CSF is also found to be involved in macrophage generation and immune regulation; however, its in vivo role in immune homeostasis is largely unknown. Here, we examined the role of G-CSF in dextran sulfate sodium (DSS)-induced acute colitis using G-CSF receptor-deficient (G-CSFR(-/-)) mice. Mice were administered with 1.5% DSS in drinking water for 5days, and the severity of colitis was measured for the next 5days. GCSFR(-/-) mice were more susceptible to DSS-induced colitis than G-CSFR(+/+) or G-CSFR(-/+) mice. G-CSFR(-/-) mice harbored less F4/80(+) macrophages, but a similar number of neutrophils, in the intestine. In vitro, bone marrow-derived macrophages prepared in the presence of both G-CSF and macrophage colony-stimulating factor (M-CSF) (G-BMDM) expressed higher levels of regulatory macrophage markers such as programmed death ligand 2 (PDL2), CD71 and CD206, but not in arginase I, transforming growth factor (TGF)-ß, Ym1 (chitinase-like 3) and FIZZ1 (found in inflammatory zone 1), and lower levels of inducible nitric oxide synthase (iNOS), CD80 and CD86 than bone marrow-derived macrophages prepared in the presence of M-CSF alone (BMDM), in response to interleukin (IL)-4/IL-13 and lipopolysaccharide (LPS)/interferon (IFN)-γ, respectively. Adoptive transfer of G-BMDM, but not BMDM, protected G-CSFR(-/-) mice from DSS-induced colitis, and suppressed expression of tumor necrosis factor (TNF)-α, IL-1ß and iNOS in the intestine. These results suggest that G-CSF plays an important role in preventing colitis, likely through populating immune regulatory macrophages in the intestine.


Subject(s)
Colitis/immunology , Colitis/prevention & control , Granulocyte Colony-Stimulating Factor/physiology , Homeostasis , Intestines/immunology , Macrophages/physiology , Adoptive Transfer , Animals , Cells, Cultured , Colitis/chemically induced , Dextran Sulfate , Interleukin-13/immunology , Interleukin-1beta/metabolism , Intestines/cytology , Intestines/physiology , Lipopolysaccharides/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Mice , Nitric Oxide Synthase Type II/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Theriogenology ; 84(7): 1075-87, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26194698

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is required for proliferation, differentiation, and survival of cells. It is also a biomarker of human oocyte developmental competence for embryo implantation. In humans, the G-CSF concentration peaks during the ovulatory phase of the ovarian cycle. In this study, the expressions of G-CSF and its receptor were analyzed by polymerase chain reaction in granulosa cells (GCs), CL, cumulus cells (CCs), and oocytes. Cumulus-oocyte complexes were aspirated from antral follicles of 1 to 3 mm (small follicles) and 4 to 6 mm (medium follicles). Cumulus-oocyte complexes from two kinds of follicles were matured in protein-free maturation medium supplemented with various concentrations of G-CSF (0, 10, and 100 ng/mL). By real-time polymerase chain reaction, the expressions of G-CSF and its receptor were detected in GCs, CL, CCs, and oocytes. Interestingly, the G-CSF transcript levels were significantly lower in oocytes than in the other cell types, whereas the G-CSF receptor transcript levels in oocytes were similar to those in GCs. After 44 hours of IVM, no differences in the rate of nuclear maturation were detected; however, the intracellular reactive oxygen species levels in oocytes from both groups of follicles matured with 10 ng/mL of human recombinant G-CSF (hrG-CSF) groups were significantly lower (P < 0.05). After parthenogenetic activation, the cleavage rates were significantly (P < 0.05) higher in 100 ng/mL hrG-CSF-treated small (63.3%) follicles than in 0, 10 ng/mL hrG-CSF-treated small (38.6% and 49.0%, respectively) follicles and 0 ng/mL hrG-CSF-treated medium (52.1%) follicles, and the cleavage rates were significantly (P < 0.05) higher in 10 ng/mL hrG-CSF-treated medium (76.3%) follicles than in all other groups. The blastocyst formation rates were significantly (P < 0.05) higher in 100 ng/mL hrG-CSF-treated small (31.2%) follicles than in 0 and 10 ng/mL hrG-CSF small (10.4% and 15.6%, respectively) follicles, and the 10 ng/mL hrG-CSF medium (45.7%) follicle was significantly (P < 0.05) higher than in all other groups. The total cell number in blastocysts from the 10 ng/mL hrG-CSF medium (106.5) follicles was significantly (P < 0.05) increased compared to 0, 10, 100 ng/mL hrG-CSF small (55.0, 73.7 and 59.5, respectively) follicles and 0, 100 ng/mL hrG-CSF-treated medium (82.5 and 93.5, respectively) follicles. After IVF, the blastocysts stage was significantly (P < 0.05) increased in 10 ng/mL hrG-CSF-treated medium (36.4%) follicles. Fertilization efficiency was significantly high in 100 ng/mL of small (29.1%) and 10 ng/mL of medium (44.0%) follicles. We also examined the Bcl2 and ERK2 transcript levels and found that they were significantly higher in the small and medium follicle treatment groups. In conclusion, these results indicate that hrG-CSF improve the viability of porcine embryos.


Subject(s)
Embryonic Development/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , In Vitro Oocyte Maturation Techniques/veterinary , Swine , Animals , Blastocyst/physiology , Cumulus Cells/chemistry , Female , Gene Expression , Granulocyte Colony-Stimulating Factor/analysis , Granulocyte Colony-Stimulating Factor/genetics , Granulosa Cells/chemistry , Humans , Oocytes/chemistry , RNA, Messenger/analysis , Reactive Oxygen Species/analysis , Real-Time Polymerase Chain Reaction , Receptors, Granulocyte Colony-Stimulating Factor/analysis , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL