Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Eur J Med Chem ; 268: 116253, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38401188

ABSTRACT

This study explores the potential of CSF-1R inhibitors as therapeutic agents for neurodegenerative diseases. CSF-1R, a receptor tyrosine kinase primarily expressed in macrophage lineages, plays a pivotal role in regulating various cellular processes. Recent research highlights the significance of CSF-1R inhibition in mitigating neuroinflammation, particularly in Alzheimer's disease, where microglial overactivation contributes to neurodegeneration. The research reveals a series of N-(5-amido-2-methylphenyl)-5-methylisoxazole-3-carboxamide CSF-1R inhibitors, where compounds 7d, 7e, and 9a exhibit outstanding inhibitory activities and selectivity, with IC50 values of 33, 31, and 64 nM, respectively. These most promising compounds in this series were profiled for cellular potency and subjected to in vitro pharmacokinetic profiling. These inhibitors exhibit minimal cytotoxicity, even at higher concentrations, and possess promising blood-brain barrier permeability, making them potential candidates for central nervous system diseases. The investigation into the in vitro ADME properties, including plasma and microsomal stability, reveals that these CSF-1R inhibitors maintain their structural integrity and plasma concentration. This resilience positions them for further development as therapeutic agents for neurodegenerative diseases.


Subject(s)
Isoxazoles , Neurodegenerative Diseases , Receptor, Macrophage Colony-Stimulating Factor , Humans , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Blood-Brain Barrier/metabolism , Receptor Protein-Tyrosine Kinases , Enzyme Inhibitors
2.
Adv Mater ; 36(19): e2310876, 2024 May.
Article in English | MEDLINE | ID: mdl-38321645

ABSTRACT

Structural and physiological cues provide guidance for the directional migration and spatial organization of endogenous cells. Here, a microchannel scaffold with instructive niches is developed using a circumferential freeze-casting technique with an alkaline salting-out strategy. Thereinto, polydopamine-coated nano-hydroxyapatite is employed as a functional inorganic linker to participate in the entanglement and crystallization of chitosan molecules. This scaffold orchestrates the advantage of an oriented porous structure for rapid cell infiltration and satisfactory immunomodulatory capacity to promote stem cell recruitment, retention, and subsequent osteogenic differentiation. Transcriptomic analysis as well as its in vitro and in vivo verification demonstrates that essential colony-stimulating factor-1 (CSF-1) factor is induced by this scaffold, and effectively bound to the target colony-stimulating factor-1 receptor (CSF-1R) on the macrophage surface to activate the M2 phenotype, achieving substantial endogenous bone regeneration. This strategy provides a simple and efficient approach for engineering inducible bone regenerative biomaterials.


Subject(s)
Bone Regeneration , Durapatite , Macrophage Colony-Stimulating Factor , Osteogenesis , Polymers , Receptor, Macrophage Colony-Stimulating Factor , Tissue Scaffolds , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Animals , Mice , Durapatite/chemistry , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Polymers/chemistry , Cell Differentiation , Chitosan/chemistry , Indoles/chemistry , Signal Transduction , Tissue Engineering/methods , Macrophages/metabolism , Macrophages/cytology , RAW 264.7 Cells
3.
Exp Mol Med ; 52(8): 1239-1254, 2020 08.
Article in English | MEDLINE | ID: mdl-32801364

ABSTRACT

Colony-stimulating factor 1 receptor (CSF1R, also known as c-FMS) is a receptor tyrosine kinase. Macrophage colony-stimulating factor (M-CSF) and IL-34 are ligands of CSF1R. CSF1R-mediated signaling is crucial for the survival, function, proliferation, and differentiation of myeloid lineage cells, including osteoclasts, monocytes/macrophages, microglia, Langerhans cells in the skin, and Paneth cells in the intestine. CSF1R also plays an important role in oocytes and trophoblastic cells in the female reproductive tract and in the maintenance and maturation of neural progenitor cells. Given that CSF1R is expressed in a wide range of myeloid cells, altered CSF1R signaling is implicated in inflammatory, neoplastic, and neurodegenerative diseases. Inhibiting CSF1R signaling through an inhibitory anti-CSF1R antibody or small molecule inhibitors that target the kinase activity of CSF1R has thus been a promising therapeutic strategy for those diseases. In this review, we cover the recent progress in our understanding of the various roles of CSF1R in osteoclasts and other myeloid cells, highlighting the therapeutic applications of CSF1R inhibitors in disease conditions.


Subject(s)
Osteoclasts/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Animals , Disease , Humans , Ligands , Models, Biological , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Signal Transduction
4.
PLoS Biol ; 16(8): e2002979, 2018 08.
Article in English | MEDLINE | ID: mdl-30142160

ABSTRACT

There is currently a demand for new highly efficient and specific drugs to treat osteoporosis, a chronic bone disease affecting millions of people worldwide. We have developed a combinatorial strategy for engineering bispecific inhibitors that simultaneously target the unique combination of c-FMS and αvß3 integrin, which act in concert to facilitate bone resorption by osteoclasts. Using functional fluorescence-activated cell sorting (FACS)-based screening assays of random mutagenesis macrophage colony-stimulating factor (M-CSF) libraries against c-FMS and αvß3 integrin, we engineered dual-specific M-CSF mutants with high affinity to both receptors. These bispecific mutants act as functional antagonists of c-FMS and αvß3 integrin activation and hence of osteoclast differentiation in vitro and osteoclast activity in vivo. This study thus introduces a versatile platform for the creation of new-generation therapeutics with high efficacy and specificity for osteoporosis and other bone diseases. It also provides new tools for studying molecular mechanisms and the cell signaling pathways that mediate osteoclast differentiation and function.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Integrin alphaVbeta3/antagonists & inhibitors , Macrophage Colony-Stimulating Factor/pharmacology , Osteoclasts/drug effects , Osteoporosis/drug therapy , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Binding Sites , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation , Humans , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Macrophage Colony-Stimulating Factor/chemistry , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Molecular Docking Simulation , Mutation , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Engineering , Protein Interaction Domains and Motifs , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
5.
J Med Chem ; 61(13): 5450-5466, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29293000

ABSTRACT

Colony stimulation factor-1 receptor (CSF-1R), which is also known as FMS kinase, plays an important role in initiating inflammatory, cancer, and bone disorders when it is overstimulated by its ligand, CSF-1. Innate immunity, as well as macrophage differentiation and survival, are regulated by the stimulation of the CSF-1R. Another ligand, interlukin-34 (IL-34), was recently reported to activate the CSF-1R receptor in a different manner. The relationship between CSF-1R and microglia has been reviewed. Both CSF-1 antibodies and small molecule CSF-1R kinase inhibitors have now been tested in animal models and in humans. In this Perspective, we discuss the role of CSF-1 and IL-34 in producing cancer, bone disorders, and inflammation. We also review the newly discovered and improved small molecule kinase inhibitors and monoclonal antibodies that have shown potent activity toward CSF-1R, reported from 2012 until 2017.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Humans , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism
6.
PLoS One ; 11(7): e0160165, 2016.
Article in English | MEDLINE | ID: mdl-27467080

ABSTRACT

The receptors tyrosine kinases (RTKs) for the colony stimulating factor-1, CSF-1R, and for the stem cell factor, SCFR or KIT, are important mediators of signal transduction. The abnormal function of these receptors, promoted by gain-of-function mutations, leads to their constitutive activation, associated with cancer or other proliferative diseases. A secondary effect of the mutations is the alteration of receptors' sensitivity to tyrosine kinase inhibitors, compromising effectiveness of these molecules in clinical treatment. In particular, the mutation V560G in KIT increases its sensitivity to Imatinib, while the D816V in KIT, and D802V in CSF-1R, triggers resistance to the drug. We analyzed the Imatinib binding affinity to the native and mutated KIT (mutations V560G, S628N and D816V) and CSF-1R (mutation D802V) by using molecular dynamics simulations and energy calculations of Imatinib•target complexes. Further, we evaluated the sensitivity of the studied KIT receptors to Imatinib by measuring the inhibition of KIT phosphorylation. Our study showed that (i) the binding free energy of Imatinib to the targets is highly correlated with their experimentally measured sensitivity; (ii) the electrostatic interactions are a decisive factor affecting the binding energy; (iii) the most deleterious impact to the Imatinib sensitivity is promoted by D802V (CSF-1R) and D816V (KIT) mutations; (iv) the role of the juxtamembrane region, JMR, in the imatinib binding is accessory. These findings contribute to a better description of the mutation-induced effects alternating the targets sensitivity to Imatinib.


Subject(s)
Mutation , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Stem Cell Factor/metabolism , Animals , COS Cells , Chlorocebus aethiops , Hydrogen Bonding , Imatinib Mesylate/chemistry , Imatinib Mesylate/metabolism , Molecular Dynamics Simulation , Protein Binding , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Stem Cell Factor/chemistry , Stem Cell Factor/genetics
7.
Biochem Soc Trans ; 44(2): 333-41, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27068937

ABSTRACT

Cancer cells employ a variety of mechanisms to evade apoptosis and senescence. Pre-eminent among these is the aberrant co-expression of growth factors and their ligands, forming an autocrine growth loop that promotes tumour formation and progression. One growth loop whose transforming potential has been repeatedly demonstrated is the CSF-1/CSF-1R axis. Expression of CSF-1 and/or CSF-1R has been documented in a number of human malignancies, including breast, prostate and ovarian cancer and classical Hodgkin's lymphoma (cHL). This review summarizes the large body of work undertaken to study the role of this cytokine receptor system in malignant transformation. These studies have attributed a key role to the CSF-1/CSF-1R axis in supporting tumour cell survival, proliferation and enhanced motility. Moreover, increasing evidence implicates paracrine interactions between CSF-1 and its receptor in defining a tumour-permissive and immunosuppressive tumour-associated stroma. Against this background, we briefly consider the prospects for therapeutic targeting of this system in malignant disease.


Subject(s)
Colony-Stimulating Factors/physiology , Neoplasms/physiopathology , Receptor, Macrophage Colony-Stimulating Factor/physiology , Humans , Ligands , Receptor, Macrophage Colony-Stimulating Factor/chemistry
8.
Dongwuxue Yanjiu ; 37(2): 96-102, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-27029867

ABSTRACT

Colony-stimulating factor 1 receptor (CSF-1R) is an important regulator of monocytes/macrophages (MO/MΦ). Although several CSF-1R genes have been identified in teleosts, the precise role of CSF- 1R in ayu (Plecoglossus altivelis) remains unclear. In this study, we characterized the CSF-1R homologue from P. altivelis, and named it PaCSF-1R. Multiple sequence alignment and phylogenetic tree analysis showed that PaCSF-1R was most closely related to that of Japanese ricefish (Oryzias latipes). Tissue distribution and expression analysis showed that the PaCSF-1R transcript was mainly expressed in the head kidney-derived MO/MΦ, spleen, and head kidney, and its expression was significantly altered in various tissues upon Vibrio anguillarum infection. After PaCSF-1R neutralization for 48 h, the phagocytic activity of MO/MΦ was significantly decreased, suggesting that PaCSF-1R plays a role in regulating the phagocytic function of ayu MO/MΦ.


Subject(s)
Fish Proteins/genetics , Gene Expression Regulation , Osmeriformes/genetics , Osmeriformes/microbiology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Vibrio/physiology , Amino Acid Sequence , Animals , Cloning, Molecular , Fish Proteins/chemistry , Fish Proteins/metabolism , Phagocytosis , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism
9.
Fish Shellfish Immunol ; 50: 223-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26828262

ABSTRACT

Macrophage colony-stimulating factor receptor (M-CSFR), a member of the group of type III protein tyrosine kinase receptors, is expressed primarily by monocyte/macrophage lineage cells. In order to describe the distribution of macrophages at the maternal-fetal interface in Neoditrema ransonnetii, a viviparous fish species, M-CSFR cDNA was sequenced. Two sequences were obtained: NrM-CSFR1 (4381 bp, encoding 980 amino acids), and NrM-CSFR2 (3573 bp, encoding 1016 amino acids). Both the genes were expressed in the ovary of pregnant females. In situ hybridization revealed that a number of cells that were positive for NrM-CSFR1 and/or NrM-CSFR2 populated the ovigerous lamellae of the ovary during pregnancy. Following parturition, M-CSFR-positive cells disappeared from the subepithelial region of ovigerous lamellae, and were localized in perivascular tissues. These results suggest the role of M-CSFR-positive cells, which appear to be macrophages, in N. ransonnetii during pregnancy.


Subject(s)
Fish Proteins/genetics , Macrophages/metabolism , Ovary/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Female , Fish Proteins/chemistry , Fish Proteins/metabolism , Perciformes , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sequence Alignment/veterinary , Viviparity, Nonmammalian
10.
Sci Signal ; 8(405): rs13, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26628682

ABSTRACT

Protein kinase autophosphorylation is a common regulatory mechanism in cell signaling pathways. Crystal structures of several homomeric protein kinase complexes have a serine, threonine, or tyrosine autophosphorylation site of one kinase monomer located in the active site of another monomer, a structural complex that we call an "autophosphorylation complex." We developed and applied a structural bioinformatics method to identify all such autophosphorylation complexes in x-ray crystallographic structures in the Protein Data Bank (PDB). We identified 15 autophosphorylation complexes in the PDB, of which five complexes had not previously been described in the publications describing the crystal structures. These five complexes consist of tyrosine residues in the N-terminal juxtamembrane regions of colony-stimulating factor 1 receptor (CSF1R, Tyr(561)) and ephrin receptor A2 (EPHA2, Tyr(594)), tyrosine residues in the activation loops of the SRC kinase family member LCK (Tyr(394)) and insulin-like growth factor 1 receptor (IGF1R, Tyr(1166)), and a serine in a nuclear localization signal region of CDC-like kinase 2 (CLK2, Ser(142)). Mutations in the complex interface may alter autophosphorylation activity and contribute to disease; therefore, we mutated residues in the autophosphorylation complex interface of LCK and found that two mutations impaired autophosphorylation (T445V and N446A) and mutation of Pro(447) to Ala, Gly, or Leu increased autophosphorylation. The identified autophosphorylation sites are conserved in many kinases, suggesting that, by homology, these complexes may provide insight into autophosphorylation complex interfaces of kinases that are relevant drug targets.


Subject(s)
Databases, Protein , Lymphocyte Specific Protein Tyrosine Kinase p56(lck) , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Receptor, EphA2 , Receptor, Macrophage Colony-Stimulating Factor , Amino Acid Substitution , HEK293 Cells , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mutation, Missense , Phosphorylation/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptor, EphA2/chemistry , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism
11.
Structure ; 23(9): 1621-1631, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26235028

ABSTRACT

Human colony-stimulating factor 1 receptor (hCSF-1R) is unique among the hematopoietic receptors because it is activated by two distinct cytokines, CSF-1 and interleukin-34 (IL-34). Despite ever-growing insights into the central role of hCSF-1R signaling in innate and adaptive immunity, inflammatory diseases, and cancer, the structural basis of the functional dichotomy of hCSF-1R has remained elusive. Here, we report crystal structures of ternary complexes between hCSF-1 and hCSF-1R, including their complete extracellular assembly, and propose a mechanism for the cooperative human CSF-1:CSF-1R complex that relies on the adoption by dimeric hCSF-1 of an active conformational state and homotypic receptor interactions. Furthermore, we trace the cytokine-binding duality of hCSF-1R to a limited set of conserved interactions mediated by functionally equivalent residues on CSF-1 and IL-34 that play into the geometric requirements of hCSF-1R activation, and map the possible mechanistic consequences of somatic mutations in hCSF-1R associated with cancer.


Subject(s)
Crystallography, X-Ray , Macrophage Colony-Stimulating Factor/chemistry , Macrophage Colony-Stimulating Factor/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Binding Sites , Enzyme Activation , Humans , Models, Molecular , Phosphorylation , Scattering, Small Angle , Signal Transduction , X-Ray Diffraction
12.
Xenobiotica ; 45(2): 107-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25180976

ABSTRACT

1. 4-[2((1R,2R)-2-Hydroxycyclohexylamino)-benzothiazol-6-yloxyl]-pyridine-2-carboxylic acid methylamide (BLZ945) is a small molecule inhibitor of CSF-1R kinase activity within osteoclasts designed to prevent skeletal related events in metastatic disease. Key metabolites were enzymatically and structurally characterized to understand the metabolic fate of BLZ945 and pharmacological implications. The relative intrinsic clearances for metabolites were derived from in vitro studies using human hepatocytes, microsomes and phenotyped with recombinant P450 enzymes. 2. Formation of a pharmacologically active metabolite (M9) was observed in human hepatocytes. The M9 metabolite is a structural isomer (diastereomer) of BLZ945 and is about 4-fold less potent. This isomer was enzymatically formed via P450 oxidation of the BLZ945 hydroxyl group, followed by aldo-keto reduction to the alcohol (M9). 3. Two reaction phenotyping approaches based on fractional clearances were applied to BLZ945 using hepatocytes and liver microsomes. The fraction metabolized (fm) or contribution ratio was determined for each metabolic reaction type (oxidation, glucuronidation or isomerization) as well as for each metabolite. The results quantitatively illustrate contribution ratios of the involved enzymes and pathways, e.g. the isomerization to metabolite M9 accounted for 24% intrinsic clearance in human hepatocytes. In summary, contribution ratios for the Phase I and Phase II pathways can be determined in hepatocytes.


Subject(s)
Benzothiazoles/metabolism , Hepatocytes/enzymology , Microsomes, Liver/enzymology , Picolinic Acids/metabolism , Protein Kinase Inhibitors/metabolism , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Cell Line , Cell Proliferation/drug effects , Cytochrome P-450 Enzyme System/metabolism , Humans , Metabolic Networks and Pathways , Picolinic Acids/chemistry , Picolinic Acids/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects
13.
Cell Signal ; 27(2): 267-74, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25446260

ABSTRACT

SLAP (Src like adaptor protein) contains adjacent Src homology 3 (SH3) and Src homology 2 (SH2) domains closely related in sequence to that of cytoplasmic Src family tyrosine kinases. Expressed most abundantly in the immune system, SLAP function has been predominantly studied in the context of lymphocyte signaling, where it functions in the Cbl dependent downregulation of antigen receptor signaling. However, accumulating evidence suggests that SLAP plays a role in the regulation of a broad range of membrane receptors including members of the receptor tyrosine kinase (RTK) family. In this review we highlight the role of SLAP in the ubiquitin dependent regulation of type III RTKs PDGFR, CSF-1R, KIT and Flt3, as well as Eph family RTKs. SLAP appears to bind activated type III and Eph RTKs via a conserved autophosphorylated juxtamembrane tyrosine motif in an SH2-dependent manner, suggesting that SLAP is important in regulating RTK signaling.


Subject(s)
Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Humans , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Proto-Oncogene Proteins pp60(c-src)/chemistry , Proto-Oncogene Proteins pp60(c-src)/genetics , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/chemistry , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , fms-Like Tyrosine Kinase 3/chemistry , fms-Like Tyrosine Kinase 3/metabolism , src Homology Domains
14.
Fish Shellfish Immunol ; 43(1): 142-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25543034

ABSTRACT

MCSF and its well-known receptor MCSFR had been well studied in humans, regulating the differentiation, proliferation, and survival of the mononuclear phagocyte system. IL-34, which is an alternative ligand of MCSF receptor, was recently identified as a novel cytokine and functionally overlaps with MCSF. However, the functional study of these receptors and their ligands in fish are largely unknown. In the present study, the cDNA of two potential grouper MCSFR ligands have been cloned, EcIL-34 (657 bp) and EcMCSF2 (804 bp), as well as an additional copy of grouper MCSFR, EcMCSFR2 (3141 bp). Sequence analysis showed that these three molecules had higher identities with other fish counterparts compared to mammals and their conserved structures and important functional residues were also analyzed. Tissue distribution analysis showed that EcIL-34 is dominant in brain, gill and spleen compared to EcMCSF2, which is dominant in head kidney, trunk kidney, skin, heart and muscle. EcMCSFR1 was dominant in the most tissues except head kidney and liver compared to EcMCSFR2. The different tissue distribution patterns of these two grouper MCSF receptors and their two ligands indicate the different mononuclear phagocyte differentiation and activation modes in different tissues. In Cryptocaryon irritans infected grouper, EcIL-34 and EcMCSFR2 were the most strongly up-regulated ligand and receptor in the infected sites, gill and skin. Their up-regulation confirmed the proliferation and activation of phagocytes in C. irritans infected sites, which would improve the antigen presentation and elicit the host local specific immune response. In C. irritans infected grouper head kidney, both ligands EcIL-34 and EcMCSF2 (especially EcMCSF2) were up-regulated, but both receptors EcMCSFR1 and EcMCSFR2 were down-regulated, which indicated that the phagocytes differentiation and proliferation may have occurred in this hemopoietic organ, and after that they migrated to the infected cites. The down-regulation of EcIL-34 and EcMCSF2 and no significant change of EcMCSFR1 and EcMCSFR2 in most time point of grouper spleen showed it was less involved in phagocytes response to C. irritans infection.


Subject(s)
Bass/genetics , Ciliophora Infections/veterinary , Fish Diseases/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Gene Expression Regulation , Amino Acid Sequence , Animals , Bass/immunology , Bass/metabolism , Ciliophora/physiology , Ciliophora Infections/genetics , Ciliophora Infections/immunology , Ciliophora Infections/parasitology , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/metabolism , Fish Diseases/parasitology , Fish Proteins/chemistry , Fish Proteins/metabolism , Interleukins/chemistry , Interleukins/genetics , Interleukins/metabolism , Macrophage Colony-Stimulating Factor/chemistry , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Organ Specificity , Phagocytes , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sequence Alignment/veterinary
15.
Cell Mol Life Sci ; 71(19): 3627-31, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24972636

ABSTRACT

Receptor tyrosine kinases (RTK) have long being studied with respect to the "canonical" signaling. This includes ligand-induced activation of a receptor tyrosine kinase at the cell surface that leads to receptor dimerization, followed by its phosphorylation in the intracellular domain and activation. The activated receptor then recruits cytoplasmic signaling molecules including other kinases. Activation of the downstream signaling cascade frequently leads to changes in gene expression following nuclear translocation of downstream targets. However, RTK themselves may localize within the nucleus, as either full-length molecules or cleaved fragments, with or without their ligands. Significant differences in this mechanism have been reported depending on the individual RTK, cellular context or disease. Accumulating evidences indicate that the colony-stimulating factor-1 receptor (CSF-1R) may localize within the nucleus. To date, however, little is known about the mechanism of CSF-1R nuclear shuttling, as well as the functional role of nuclear CSF-1R.


Subject(s)
Cell Nucleus/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Active Transport, Cell Nucleus , Animals , Humans , Ligands , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mice , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Signal Transduction
16.
Biochem Biophys Res Commun ; 450(1): 782-7, 2014 Jul 18.
Article in English | MEDLINE | ID: mdl-24955855

ABSTRACT

Stimulation of macrophages with phorbolesters, bacterial DNA, or lipopolysaccharides causes regulated intramembrane proteolysis or RIPping of the CSF-1 receptor. This process involves TACE-mediated cleavage in the extracellular domain, followed by γ-secretase-mediated cleavage within the transmembrane region. In the current study, we have identified the TACE cleavage site, which is present twelve residues from the carboxy-terminal end of the extracellular domain. Replacement of fourteen residues at the end of the extracellular domain blocked TACE cleavage. In addition, we identified the γ-secretase cleavage site, which is present four residues from the carboxy-terminal end of the transmembrane region. Replacement of six residues surrounding this site strongly reduced intramembrane cleavage. Our results provide new insights into the molecular physiology of the CSF-1 receptor and contribute to our understanding of substrate selection by TACE and γ-secretase.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/metabolism , ADAM Proteins/genetics , ADAM17 Protein , Amyloid Precursor Protein Secretases/genetics , Binding Sites , Enzyme Activation , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Protein Binding , Receptor, Macrophage Colony-Stimulating Factor/genetics
17.
Article in English | MEDLINE | ID: mdl-24890514

ABSTRACT

The CSF-1 receptor (CSF-1R) is activated by the homodimeric growth factors colony-stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). It plays important roles in development and in innate immunity by regulating the development of most tissue macrophages and osteoclasts, of Langerhans cells of the skin, of Paneth cells of the small intestine, and of brain microglia. It also regulates the differentiation of neural progenitor cells and controls functions of oocytes and trophoblastic cells in the female reproductive tract. Owing to this broad tissue expression pattern, it plays a central role in neoplastic, inflammatory, and neurological diseases. In this review we summarize the evolution, structure, and regulation of expression of the CSF-1R gene. We discuss the structures of CSF-1, IL-34, and the CSF-1R and the mechanism of ligand binding to and activation of the receptor. We further describe the pathways regulating macrophage survival, proliferation, differentiation, and chemotaxis downstream from the CSF-1R.


Subject(s)
Myeloid Cells/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Animals , Evolution, Molecular , Humans , Ligands , Myeloid Cells/cytology , Protein Conformation , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics
18.
PLoS One ; 9(5): e97519, 2014.
Article in English | MEDLINE | ID: mdl-24828813

ABSTRACT

The colony stimulating factor-1 receptor (CSF-1R) and the stem cell factor receptor KIT, type III receptor tyrosine kinases (RTKs), are important mediators of signal transduction. The normal functions of these receptors can be compromised by gain-of-function mutations associated with different physiopatological impacts. Whereas KIT D816V/H mutation is a well-characterized oncogenic event and principal cause of systemic mastocytosis, the homologous CSF-1R D802V has not been identified in human cancers. The KIT D816V oncogenic mutation triggers resistance to the RTK inhibitor Imatinib used as first line treatment against chronic myeloid leukemia and gastrointestinal tumors. CSF-1R is also sensitive to Imatinib and this sensitivity is altered by mutation D802V. Previous in silico characterization of the D816V mutation in KIT evidenced that the mutation caused a structure reorganization of the juxtamembrane region (JMR) and facilitated its departure from the kinase domain (KD). In this study, we showed that the equivalent CSF-1R D802V mutation does not promote such structural effects on the JMR despite of a reduction on some key H-bonds interactions controlling the JMR binding to the KD. In addition, this mutation disrupts the allosteric communication between two essential regulatory fragments of the receptors, the JMR and the A-loop. Nevertheless, the mutation-induced shift towards an active conformation observed in KIT D816V is not observed in CSF-1R D802V. The distinct impact of equivalent mutation in two homologous RTKs could be associated with the sequence difference between both receptors in the native form, particularly in the JMR region. A local mutation-induced perturbation on the A-loop structure observed in both receptors indicates the stabilization of an inactive non-inhibited form, which Imatinib cannot bind.


Subject(s)
Mutation , Proto-Oncogene Proteins c-kit/chemistry , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Allosteric Regulation , Amino Acid Sequence , Antineoplastic Agents/chemistry , Benzamides/chemistry , Eukaryotic Cells/metabolism , Eukaryotic Cells/pathology , Humans , Imatinib Mesylate , Molecular Dynamics Simulation , Molecular Sequence Data , Piperazines/chemistry , Principal Component Analysis , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/chemistry , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics
19.
J Proteome Res ; 13(3): 1428-37, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24422531

ABSTRACT

The importance of diagnosis and therapies for liver cirrhosis (LC) is indisputable. Thus, a reliable method for monitoring the progression of liver fibrosis and resultant LC is urgently needed. Previously, using a lectin-assisted glycoproteomic method, we identified 26 serum glycoproteins as promising glycobiomarker candidates for monitoring the progression of liver diseases. In this study, we identified colony stimulating factor 1 receptor (CSF1R) as a promising LC marker candidate and then established Wisteria floribunda agglutinin (WFA)-reactive CSF1R (WFA(+)-CSF1R) as a novel possible glycobiomarker candidate by utilizing a glycoproteomics-based strategy. The serum level of WFA(+)-CSF1R in patients with hepatitis C virus (HCV)-infected liver disease was measured by an antibody-lectin sandwich ELISA. In a proof-of-concept experiment of the strategy preceding to future clinical studies, LC patients showed a high serum WFA(+)-CSF1R level in selected samples (P = 1.3 × 10(-17)). This result suggests WFA(+)-CSF1R is a possible biomarker candidate for evaluation of LC. Our results verified feasibility of this strategy for glycobiomarker development.


Subject(s)
Glycoproteins/blood , Liver Cirrhosis/blood , Plant Lectins/chemistry , Polysaccharides/analysis , Receptor, Macrophage Colony-Stimulating Factor/blood , Receptors, N-Acetylglucosamine/chemistry , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Carbohydrate Conformation , Enzyme-Linked Immunosorbent Assay , Feasibility Studies , Female , Glycoproteins/chemistry , Hepatitis B, Chronic/blood , Hepatitis C, Chronic/blood , Humans , Liver Cirrhosis/diagnosis , Male , Middle Aged , Polysaccharides/chemistry , Protein Array Analysis , Proteomics , Receptor, Macrophage Colony-Stimulating Factor/chemistry
20.
Biotechnol Prog ; 30(2): 411-7, 2014.
Article in English | MEDLINE | ID: mdl-24375974

ABSTRACT

Since cell migration plays critical roles in development and homeostasis of the body, artificial control of cell migration would be promising for the treatment of various diseases related to migration. To this end, we previously developed single-chain Fv (scFv)/receptor chimeras, named signalobodies, which can control cell fates via a specific antigen that is different from natural cytokines. Although a conventional chemotaxis chamber assay revealed that several signalobodies based on receptor tyrosine kinases transduced antigen-dependent migration signals, we have never performed direct observation of the cells to obtain more information on overall properties of cell motility and migration. In this study, we utilized murine pro-B Ba/F3 cells expressing either a scFv-Fms or scFv-Mpl signalobody, and compared their migratory characteristics. We employed a lipid-polyethylene glycol conjugate to softly immobilize the suspension cells on a slide, which facilitated direct observation of chemokinetic activity of the cells. Consequently, both cells markedly exhibited chemokinesis in response to a specific antigen. In addition, the cells were subjected to a stable antigen-concentration gradient to observe horizontal directional cell migration in real time. The results showed that the cells expressing scFv-Fms underwent directional migration toward a positive antigen-concentration gradient. Taken together, we successfully demonstrated antigen-responsive regulation of cell motility and migration via the signalobodies.


Subject(s)
Cell Movement/physiology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Thrombopoietin/metabolism , Signal Transduction/physiology , Single-Chain Antibodies/metabolism , Animals , Antigens/metabolism , Cell Line , Fluorescein/chemistry , Fluorescein/metabolism , Mice , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptors, Thrombopoietin/chemistry , Single-Chain Antibodies/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...