Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 9032, 2024 04 19.
Article in English | MEDLINE | ID: mdl-38641704

ABSTRACT

CSF1R is a receptor tyrosine kinase responsible for the growth/survival/polarization of macrophages and overexpressed in some AML patients. We hypothesized that a novel multi-kinase inhibitor (TKi), narazaciclib (HX301/ON123300), with high potency against CSF1R (IC50 ~ 0.285 nM), would have anti-AML effects. We tested this by confirming HX301's high potency against CSF1R (IC50 ~ 0.285 nM), as well as other kinases, e.g. FLT3 (IC50 of ~ 19.77 nM) and CDK6 (0.53 nM). An in vitro proliferation assay showed that narazaciclib has a high growth inhibitory effect in cell cultures where CSF1R or mutant FLT3-ITD variants that may be proliferation drivers, including primary macrophages (IC50 of 72.5 nM) and a subset of AML lines (IC50 < 1.5 µM). In vivo pharmacology modeling of narazaciclib using five AML xenografts resulted in: inhibition of MV4-11 (FLT3-ITD) subcutaneous tumor growth and complete suppression of AM7577-PDX (FLT3-ITD/CSF1Rmed) systemic growth, likely due to the suppression of FLT3-ITD activity; complete suppression of AM8096-PDX (CSF1Rhi/wild-type FLT3) growth, likely due to the inhibition of CSF1R ("a putative driver"); and nonresponse of both AM5512-PDX and AM7407-PDX (wild-type FLT3/CSF1Rlo). Significant leukemia load reductions in bone marrow, where disease originated, were also achieved in both responders (AM7577/AM8096), implicating that HX301 might be a potentially more effective therapy than those only affecting peripheral leukemic cells. Altogether, narazaciclib can potentially be a candidate treatment for a subset of AML with CSF1Rhi and/or mutant FLT3-ITD variants, particularly second generation FLT3 inhibitor resistant variants.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Protein Kinase Inhibitors , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor Protein-Tyrosine Kinases , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/metabolism , Pyridones/pharmacology , Pyrimidines/pharmacology
2.
Invest New Drugs ; 39(5): 1284-1297, 2021 10.
Article in English | MEDLINE | ID: mdl-33852104

ABSTRACT

Background LY3022855 is a recombinant, immunoglobulin, human monoclonal antibody targeting the colony-stimulating factor-1 receptor. This phase 1 trial determined the safety, pharmacokinetics, and antitumor activity of LY3022855 in combination with durvalumab or tremelimumab in patients with advanced solid cancers who had received standard anti-cancer treatments. Methods In Part A (dose-escalation), patients received intravenous (IV) LY3022855 25/50/75/100 mg once weekly (QW) combined with durvalumab 750 mg once every two weeks (Q2W) IV or LY3022855 50 or 100 mg QW IV with tremelimumab 75/225/750 mg once every four weeks. In Part B (dose-expansion), patients with non-small cell lung cancer (NSCLC) or ovarian cancer (OC) received recommended phase 2 dose (RP2D) of LY3022855 from Part A and durvalumab 750 mg Q2W. Results Seventy-two patients were enrolled (median age 61 years): Part A = 33, Part B = 39. In Part A, maximum tolerated dose was not reached, and LY3022855 100 mg QW and durvalumab 750 mg Q2W was the RP2D. Four dose-limiting equivalent toxicities occurred in two patients from OC cohort. In Part A, maximum concentration, area under the concentration-time curve, and serum concentration showed dose-dependent increase over two cycles of therapy. Overall rates of complete response, partial response, and disease control were 1.4%, 2.8%, and 33.3%. Treatment-emergent anti-drug antibodies were observed in 21.2% of patients. Conclusions LY3022855 combined with durvalumab or tremelimumab in patients with advanced NSCLC or OC had limited clinical activity, was well tolerated. The RP2D was LY3022855 100 mg QW with durvalumab 750 mg Q2W. ClinicalTrials.gov ID: NCT02718911 (Registration Date: May 3, 2011).


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Ovarian Neoplasms/drug therapy , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Area Under Curve , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged
3.
Addict Biol ; 26(2): e12889, 2021 03.
Article in English | MEDLINE | ID: mdl-32176824

ABSTRACT

Alcohol abuse induces changes in microglia morphology and immune function, but whether microglia initiate or simply amplify the harmful effects of alcohol exposure is still a matter of debate. Here, we determine microglia function in acute and voluntary drinking behaviors using a colony-stimulating factor 1 receptor inhibitor (PLX5622). We show that microglia depletion does not alter the sedative or hypnotic effects of acute intoxication. Microglia depletion also does not change the escalation or maintenance of chronic voluntary alcohol consumption. Transcriptomic analysis revealed that although many immune genes have been implicated in alcohol abuse, downregulation of microglia genes does not necessitate changes in alcohol intake. Instead, microglia depletion and chronic alcohol result in compensatory upregulation of alcohol-responsive, reactive astrocyte genes, indicating astrocytes may play a role in regulation of these alcohol behaviors. Taken together, our behavioral and transcriptional data indicate that microglia are not the primary effector cell responsible for regulation of acute and voluntary alcohol behaviors. Because microglia depletion did not regulate acute or voluntary alcohol behaviors, we hypothesized that these doses were insufficient to activate microglia and recruit them to an effector phenotype. Therefore, we used a model of repeated immune activation using polyinosinic:polycytidylic acid (poly(I:C)) to activate microglia. Microglia depletion blocked poly(I:C)-induced escalations in alcohol intake, indicating microglia regulate drinking behaviors with sufficient immune activation. By testing the functional role of microglia in alcohol behaviors, we provide insight into when microglia are causal and when they are consequential for the transition from alcohol use to dependence.


Subject(s)
Alcoholism/pathology , Microglia/drug effects , Organic Chemicals/pharmacology , Alcohol Drinking/pathology , Alcoholic Intoxication/pathology , Animals , Astrocytes/drug effects , Chronic Disease , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Motor Skills/drug effects , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Signal Transduction/drug effects , Sleep/drug effects
4.
Glia ; 68(3): 574-588, 2020 03.
Article in English | MEDLINE | ID: mdl-31652020

ABSTRACT

The role of microglia in retinal inflammation is still ambiguous. Branch retinal vein occlusion initiates an inflammatory response whereby resident microglia cells are activated. They trigger infiltration of neutrophils that exacerbate blood-retina barrier damage, regulate postischemic inflammation and irreversible loss of neuroretina. Suppression of microglia-mediated inflammation might bear potential for mitigating functional impairment after retinal vein occlusion (RVO). To test this hypothesis, we depleted microglia by PLX5622 (a selective tyrosine kinase inhibitor that targets the colony-stimulating factor-1 receptor) in fractalkine receptor reporter mice (Cx3cr1gfp/+ ) subjected to various regimens of PLX5622 treatment and experimental RVO. Effectiveness of microglia suppression and retinal outcomes including retinal thickness as well as ganglion cell survival were compared to a control group of mice with experimental vein occlusion only. PLX5622 caused dramatic suppression of microglia. Despite vein occlusion, reappearance of green fluorescent protein positive cells was strongly impeded with continuous PLX5622 treatment and significantly delayed after its cessation. In depleted mice, retinal proinflammatory cytokine signaling was diminished and retinal ganglion cell survival improved by almost 50% compared to nondepleted animals 3 weeks after vein occlusion. Optical coherence tomography suggested delayed retinal degeneration in depleted mice. In summary, findings indicate that suppression of cells bearing the colony-stimulating factor-1 receptor, mainly microglia and monocytes, mitigates ischemic damage and salvages retinal ganglion cells. Blood-retina barrier breakdown seems central in the disease mechanism, and complex interactions between different cell types composing the blood-retina barrier as well as sustained hypoxia might explain why the protective effect was only partial.


Subject(s)
Inflammation/metabolism , Retina/pathology , Retinal Degeneration/pathology , Retinal Vein Occlusion/pathology , Animals , Blood-Retinal Barrier/pathology , Cytokines/metabolism , Disease Models, Animal , Macrophages/metabolism , Mice , Microglia/metabolism , Microglia/pathology , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/pathology , Retinal Vein Occlusion/metabolism
5.
Sci Rep ; 9(1): 14551, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601938

ABSTRACT

Tenosynovial giant cell tumors (TGCT), are rare colony stimulating factor-1(CSF-1)-driven proliferative disorders affecting joints. Diffuse-type TGCT often causes significant morbidity due to local recurrences necessitating multiple surgeries. Imatinib mesylate (IM) blocks the CSF-1 receptor. This study investigated the long term effects of IM in TGCT. We conducted an international multi-institutional retrospective study to assess the activity of IM: data was collected anonymously from individual patients with locally advanced, recurrent or metastatic TGCT. Sixty-two patients from 12 institutions across Europe, Australia and the United States were identified. Four patients with metastatic TGCT progressed rapidly on IM and were excluded for further analyses. Seventeen of 58 evaluable patients achieved complete response (CR) or partial response (PR). One- and five-year progression-free survival rates were 71% and 48%, respectively. Thirty-eight (66%) patients discontinued IM after a median of 7 (range 1-80) months. Reported adverse events in 45 (78%) patients were among other edema (48%) and fatigue (50%), mostly grade 1-2 (89%). Five patients experienced grade 3-4 toxicities. This study confirms, with additional follow-up, the efficacy of IM in TGCT. In responding cases we confirmed prolonged IM activity on TGCT symptoms even after discontinuation, but with high rates of treatment interruption and additional treatments.


Subject(s)
Antineoplastic Agents/therapeutic use , Giant Cell Tumor of Tendon Sheath/drug therapy , Imatinib Mesylate/therapeutic use , Adult , Australia , Disease Progression , Disease-Free Survival , Europe , Female , Humans , Middle Aged , Neoplasm Metastasis , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Retrospective Studies , Treatment Outcome , United States
6.
Glia ; 67(2): 291-308, 2019 02.
Article in English | MEDLINE | ID: mdl-30456797

ABSTRACT

Multiple sclerosis (MS) is one of the most common causes of progressive disability affecting young people with very few therapeutic options available for its progressive forms. Its pathophysiology involves demyelination and neurodegeneration apparently driven by microglial activation, which is physiologically dependent on colony-stimulating factor-1 receptor (CSF-1R) signaling. In the present work, we used microglial modulation through oral administration of brain-penetrant CSF-1R inhibitor BLZ945 in acute and chronic cuprizone (CPZ)-induced demyelination to evaluate preventive and therapeutic effects on de/remyelination and neurodegeneration. Our results show that BLZ945 induced a significant reduction in the number of microglia. Preventive BLZ945 treatment attenuated demyelination in the acute CPZ model, mainly in cortex and external capsule. In contrast, BLZ945 treatment in the acute CPZ model failed to protect myelin or foster remyelination in myelin-rich areas, which may respond to a loss in microglial phagocytic capacity and the consequent impairment in oligodendroglial differentiation. Preventive and therapeutic BLZ945 treatment promoted remyelination and neuroprotection in the chronic model. These results could be potentially transferred to the treatment of progressive forms of MS.


Subject(s)
Demyelinating Diseases/metabolism , Microglia/metabolism , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/metabolism , Amyloid beta-Peptides/metabolism , Animals , Benzothiazoles/therapeutic use , Brain/drug effects , Brain/pathology , Brain/ultrastructure , Bromodeoxyuridine/metabolism , Cuprizone/toxicity , Cytokines/metabolism , Demyelinating Diseases/chemically induced , Demyelinating Diseases/drug therapy , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Microglia/ultrastructure , Microscopy, Electron, Transmission , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Picolinic Acids/therapeutic use , Receptors, Colony-Stimulating Factor/genetics , Time Factors
7.
J Neuroinflammation ; 15(1): 340, 2018 Dec 12.
Article in English | MEDLINE | ID: mdl-30541565

ABSTRACT

BACKGROUND: Microglia-associated inflammation is closely related to the pathogenesis of various retinal diseases such as uveitis and diabetic retinopathy, which are associated with increased vascular permeability. In this study, we investigated the effect of systemic lipopolysaccharide (LPS) exposure to activation and proliferation of retinal microglia /macrophages. METHODS: Balb/c and Cx3cr1gfp/+ mice were challenged with LPS (1 mg/kg) daily for four consecutive days. For microglia depletion, mice were treated with colony-stimulating factor 1 receptor (CSF-1R) inhibitor PLX5622 1 week before the first LPS challenge and until the end of the experiment. In vivo imaging of the retina was performed on days 4 and 7 after the first LPS challenge, using optical coherence tomography and fluorescein angiography. Flow cytometry analysis, retinal whole mount, and retinal sections were used to investigate microglia and macrophage infiltration and proliferation after LPS challenge. Cytokines were analyzed in the blood as well as in the retina. Data analysis was performed using unpaired t tests, repeated measures one-way ANOVA, or ordinary one-way ANOVA followed by Tukey's post hoc analysis. Kruskal-Wallis test followed by Dunn's multiple comparison tests was used for the analysis of non-normally distributed data. RESULTS: Repeated LPS challenge led to activation and proliferation of retinal microglia, infiltration of monocyte-derived macrophages into the retina, and breakdown of the blood-retina barrier (BRB) accompanied by accumulation of sub-retinal fluid. Using in vivo imaging, we show that the breakdown of the BRB is highly reproducible but transitory. Acute but not chronic systemic exposure to LPS triggered a robust release of inflammatory mediators in the retina with minimal effects in the blood plasma. Inhibition of the CSF-1R by PLX5622 resulted in depletion of retinal microglia, suppression of cytokine production in the retina, and prevention of BRB breakdown. CONCLUSIONS: These findings suggest that microglia/macrophages play an important role in the pathology of retinal disorders characterized by breakdown of the BRB, and suppression of their activation may be a potential therapeutic target for such retinopathies.


Subject(s)
Blood-Retinal Barrier/drug effects , Cytokines/metabolism , Inflammation/pathology , Organic Chemicals/pharmacology , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/metabolism , Retina/pathology , Animals , Blood-Retinal Barrier/pathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cell Movement/drug effects , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inflammation/chemically induced , Ki-67 Antigen/metabolism , Lectins/metabolism , Lipopolysaccharides/toxicity , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Retina/drug effects , Retina/metabolism , Time Factors , Tomography, Optical Coherence
8.
Sci Rep ; 7(1): 17525, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235509

ABSTRACT

Complex alterations in cerebral energetic metabolism arise after traumatic brain injury (TBI). To date, methods allowing for metabolic evaluation are highly invasive, limiting our understanding of metabolic impairments associated with TBI pathogenesis. We investigated whether 13C MRSI of hyperpolarized (HP) [1-13C] pyruvate, a non-invasive metabolic imaging method, could detect metabolic changes in controlled cortical injury (CCI) mice (n = 57). Our results show that HP [1-13C] lactate-to-pyruvate ratios were increased in the injured cortex at acute (12/24 hours) and sub-acute (7 days) time points after injury, in line with decreased pyruvate dehydrogenase (PDH) activity, suggesting impairment of the oxidative phosphorylation pathway. We then used the colony-stimulating factor-1 receptor inhibitor PLX5622 to deplete brain resident microglia prior to and after CCI, in order to confirm that modulations of HP [1-13C] lactate-to-pyruvate ratios were linked to microglial activation. Despite CCI, the HP [1-13C] lactate-to-pyruvate ratio at the injury cortex of microglia-depleted animals at 7 days post-injury remained unchanged compared to contralateral hemisphere, and PDH activity was not affected. Altogether, our results demonstrate that HP [1-13C] pyruvate has great potential for in vivo non-invasive detection of cerebral metabolism post-TBI, providing a new tool to monitor the effect of therapies targeting microglia/macrophages activation after TBI.


Subject(s)
Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Brain/diagnostic imaging , Brain/metabolism , Magnetic Resonance Imaging/methods , Animals , Brain/drug effects , Carbon Isotopes , Disease Models, Animal , Lactic Acid/metabolism , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Organic Chemicals/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyruvic Acid/metabolism , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/metabolism , Spectrophotometry , Superior Sagittal Sinus , Time Factors
9.
Eur J Med Chem ; 141: 657-675, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29107425

ABSTRACT

Tyrosine kinases including LCK and FMS are involved in inflammatory disorders as well as many types of cancer. Our team has designed and synthesized thirty novel pyrimidine based inhibitors targeting LCK, classified into four different series (amides, ureas, imines (Schiff base) and benzylamines). Twelve of them showed nanomolar IC50 values. Compound 7g showed excellent selectivity profile and was selectively potent over FMS kinase (IC50 value of 4.6 nM). Molecular docking study was performed to help us rationalize the obtained results and predict the possible binding mode for our compounds in both LCK and FMS. Based on the obtained biological assay data and modelling results, a detailed SAR study was discussed. As a further testing regarding the anti-inflammatory effect of the new compounds, in vitro cellular assay over RAW 264.7 macrophages was performed. Compound 7g exhibited excellent anti-inflammatory effect. Therefore, we report the design of novel phenoxypyrimidine derivatives as potent and selective LCK inhibitors and the discovery of 7g as potent and selective FMS/LCK dual inhibitor for the potential application in inflammatory disorders including rheumatoid arthritis (RA).


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/drug therapy , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Inflammation/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , RAW 264.7 Cells , Structure-Activity Relationship
10.
J Neurosci ; 37(25): 6113-6124, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28539419

ABSTRACT

The role of microglia in degenerative and regenerative processes after damage of the nervous system remains ambiguous, partially due to the paucity of appropriate investigative methods. Here, we show that treatment with the pharmacological colony stimulating factor 1 receptor inhibitor PLX5622 specifically eliminated microglia in murine retinae and optic nerves with high efficiency. Interestingly, time course and extent of retinal ganglion cell (RGC) degeneration after optic nerve crush remained unaffected upon microglia depletion, although remnants of prelabeled apoptotic RGCs were not cleared from the retina in these animals. In addition, microglia depletion neither affected the induction of regeneration associated genes upon optic nerve injury nor the increased regenerative potential of RGCs upon lens injury (LI). However, although the repopulation of the optic nerve lesion site by astrocytes was significantly delayed upon microglia depletion, spontaneous and LI-induced axon regeneration were unaffected by PLX5622 treatment or peripheral macrophage depletion by clodronate liposome treatment. Only concurrent double depletion of microglia and infiltrated macrophages slightly, but significantly, compromised optic nerve regeneration. Therefore, microglia are not essentially involved in RGC degeneration or axonal regeneration after acute CNS injury.SIGNIFICANCE STATEMENT The roles of microglia, the phagocytosing cells of the CNS, and invading macrophages in degenerative and regenerative processes after injury are still controversial and insufficiently characterized. Here, we show that application of a CSF1R inhibitor eliminated virtually all microglia from the visual system, whereas macrophages were spared. Specific microglia depletion impaired the removal of dead labeled retinal ganglion cells after optic nerve crush, but remarkable had no influence on their degeneration. Similarly, optic nerve regeneration was completely unaffected, although repopulation of the lesion site by astrocytes was delayed significantly. Therefore, contrary to previous reports, this experimental approach revealed that microglia seemingly neither promote nor inhibit neuronal degeneration or axonal regrowth within the injured visual system.


Subject(s)
Axons/pathology , Microglia/pathology , Nerve Degeneration/pathology , Nerve Regeneration , Optic Nerve Injuries/pathology , Animals , Astrocytes/pathology , Female , Lens, Crystalline/injuries , Lens, Crystalline/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Retina/pathology , Retinal Ganglion Cells/drug effects
11.
Sci Rep ; 7: 44455, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28281657

ABSTRACT

Granulocyte colony-stimulating factor (GCSF) and its receptor (GCSFR), also known as CSF3 and CSF3R, are required to maintain normal neutrophil numbers during basal and emergency granulopoiesis in humans, mice and zebrafish. Previous studies identified two zebrafish CSF3 ligands and a single CSF3 receptor. Transient antisense morpholino oligonucleotide knockdown of both these ligands and receptor reduces neutrophil numbers in zebrafish embryos, a technique widely used to evaluate neutrophil contributions to models of infection, inflammation and regeneration. We created an allelic series of zebrafish csf3r mutants by CRISPR/Cas9 mutagenesis targeting csf3r exon 2. Biallelic csf3r mutant embryos are viable and have normal early survival, despite a substantial reduction of their neutrophil population size, and normal macrophage abundance. Heterozygotes have a haploinsufficiency phenotype with an intermediate reduction in neutrophil numbers. csf3r mutants are viable as adults, with a 50% reduction in tissue neutrophil density and a substantial reduction in the number of myeloid cells in the kidney marrow. These csf3r mutants are a new animal model of human CSF3R-dependent congenital neutropenia. Furthermore, they will be valuable for studying the impact of neutrophil loss in the context of other zebrafish disease models by providing a genetically stable, persistent, reproducible neutrophil deficiency state throughout life.


Subject(s)
Gene Editing/methods , Granulocyte Colony-Stimulating Factor/genetics , Kidney/pathology , Neutropenia/congenital , Neutrophils/pathology , Receptors, Colony-Stimulating Factor/genetics , Animals , Base Sequence , CRISPR-Cas Systems , Congenital Bone Marrow Failure Syndromes , Disease Models, Animal , Embryo, Nonmammalian , Exons , Gene Expression , Granulocyte Colony-Stimulating Factor/immunology , Haploinsufficiency , Heterozygote , Humans , Kidney/immunology , Leukocyte Count , Morpholinos/genetics , Morpholinos/metabolism , Neutropenia/genetics , Neutropenia/immunology , Neutropenia/pathology , Neutrophils/immunology , Phenotype , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/deficiency , Receptors, Colony-Stimulating Factor/immunology , Zebrafish
12.
Cancer Res ; 76(1): 35-42, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26573801

ABSTRACT

Tumors contain a heterogeneous myeloid fraction comprised of discrete MHC-II(hi) and MHC-II(lo) tumor-associated macrophage (TAM) subpopulations that originate from Ly6C(hi) monocytes. However, the mechanisms regulating the abundance and phenotype of distinct TAM subsets remain unknown. Here, we investigated the role of macrophage colony-stimulating factor (M-CSF) in TAM differentiation and polarization in different mouse tumor models. We demonstrate that treatment of tumor-bearing mice with a blocking anti-M-CSFR monoclonal antibody resulted in a reduction of mature TAMs due to impaired recruitment, extravasation, proliferation, and maturation of their Ly6C(hi) monocytic precursors. M-CSFR signaling blockade shifted the MHC-II(lo)/MHC-II(hi) TAM balance in favor of the latter as observed by the preferential differentiation of Ly6C(hi) monocytes into MHC-II(hi) TAMs. In addition, the genetic and functional signatures of MHC-II(lo) TAMs were downregulated upon M-CSFR blockade, indicating that M-CSFR signaling shapes the MHC-II(lo) TAM phenotype. Conversely, granulocyte macrophage (GM)-CSFR had no effect on the mononuclear tumor infiltrate or relative abundance of TAM subsets. However, GM-CSFR signaling played an important role in fine-tuning the MHC-II(hi) phenotype. Overall, our data uncover the multifaceted and opposing roles of M-CSFR and GM-CSFR signaling in governing the phenotype of macrophage subsets in tumors, and provide new insight into the mechanism of action underlying M-CSFR blockade.


Subject(s)
Macrophages/metabolism , Monocytes/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Tumor Microenvironment/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cell Differentiation/physiology , Cell Polarity/physiology , Female , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Monocytes/pathology , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Signal Transduction
13.
Curr Top Med Chem ; 9(7): 599-610, 2009.
Article in English | MEDLINE | ID: mdl-19689368

ABSTRACT

FMS is the exclusive receptor tyrosine kinase for colony-stimulating factor-1 (CSF-1, also known as M-CSF), which regulates the survival, proliferation, differentiation, and function of macrophage lineage cells. Since CSF-1 is over-expressed in many tumors and at sites of inflammation, small molecule inhibitors of CSF-1 appear to offer an attractive strategy for reducing macrophage numbers associated with cancer as well as autoimmune and inflammatory disease, such as rheumatoid arthritis (RA). Numerous FMS inhibitors with structurally distinct chemotypes have been developed and exhibit potent in vitro activity, but only a limited number of compounds have progressed clinically due to poor selectivity. To date, only a handful of FMS inhibitors have been tested in models of metastatic bone disease and RA. This review will summarize the biology of FMS and its function in bone physiology, inflammation, immunity, and cancer. In addition, efforts directed towards identifying FMS-selective small molecule inhibitors as well as the advancement of non-selective inhibitors in the clinic will be highlighted. Furthermore, emerging monoclonal antibody-based therapeutic strategies specifically targeting M-CSF will be described.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Macrophage Colony-Stimulating Factor/physiology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Bone and Bones/physiology , Humans , Substrate Specificity
15.
Hematol Cell Ther ; 39(2): 98-101, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9168307

ABSTRACT

Gene inactivation data have documented that erythropoietin, G-CSF, M-CSF and thrombopoietin are major regulators in vivo respectively of red cell, neutrophil, macrophage and platelet production. Transgenic mice can provide valuable models for observing the consequences of excessive stimulation by a particular growth factor but are subject to variation based on the use of differing promoters for the inserted gene.


Subject(s)
Colony-Stimulating Factors/antagonists & inhibitors , Colony-Stimulating Factors/genetics , Gene Expression Regulation , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/genetics , Animals , Colony-Stimulating Factors/biosynthesis , Mice , Receptors, Colony-Stimulating Factor/biosynthesis
16.
Blood ; 86(7): 2590-7, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7545461

ABSTRACT

Regulation of development of hematopoietic stem cells was examined by culturing Lin- c-Kit+ Sca1+ stem cells sorted from bone marrow (BM) cells by fluorescence-activated cell sorting on a layer of TBR59, a BM stromal cell line established from simian virus 40 T-antigen gene transgenic mice. The sorted stem cells did not show self-renewal, but two waves (at 7 and 13 days) of a cobblestone formation were induced by the stromal cell layer. The cobblestones were formed by finite cell division (eight divisions on average) of sorted Lin- c-Kit+ Sca1+ stem cells, and divided cells were still immature. The c-Kithigh stem cell population was induced to form the first wave of cobblestone formation committed to myeloid lineage, whereas c-Kitlow population was induced to form the second wave of this formation committed to lymphoid lineage. Both cobblestone formations require c-Kit function, but very late activation antigen-4-vascular cell adhesion molecule-1 interaction plays different parts in the two lineages.


Subject(s)
Bone Marrow Cells , Cell Separation , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Stromal Cells/physiology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Ly/analysis , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/physiology , Cells, Cultured , Flow Cytometry , Hematopoietic Stem Cells/chemistry , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Colony-Stimulating Factor/analysis , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/physiology , Receptors, Very Late Antigen/antagonists & inhibitors , Receptors, Very Late Antigen/physiology , Vascular Cell Adhesion Molecule-1
17.
Cell Tissue Res ; 280(1): 97-111, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7538451

ABSTRACT

In vivo injection of a neutralizing, monoclonal antibody (ACK2) to the receptor tyrosine kinase (c-kit) disrupts the normal motility patterns of the mouse small intestine. Immunohistochemical studies showed that cells expressing c-kit-like immunoreactivity (c-kit-LI) decreased in numbers in response to ACK2, but the identity of these cells is unknown. We investigated the identity and development of the cells that express c-kit-LI in the mouse small intestine and colon. Cells in the region of the myenteric plexus and deep muscular plexus of the small intestine and in the subserosa, in the myenteric plexus region, within the circular and longitudinal muscle layers, and along the submucosal surface of the circular muscle in the colon were labeled with ACK2. The distribution of cells that express c-kit-LI was the same as that of interstitial cells (ICs). In whole-mount preparations cells with c-kit-LI were interconnected, forming a network similar to the network formed by cells that stained with methylene blue, which has been used as a marker for ICs in the mouse gastrointestinal tract. Immunocytochemistry verified that ICs were labeled with ACK2. Multiple injections of animals with ACK2 between days 0 and 8 post partum (pp) caused a dramatic reduction in the number of ICs compared to control animals. From an ultrastructural point of view, the proliferation and development appeared to be suppressed in some classes of ICs, while others displayed an altered course of development. Functional studies showed that the decrease in ICs was accompanied by a loss of electrical rhythmicity in the small intestine and reduced neural responses in the small bowel and colon. Morphological experiments showed that c-kit-positive cells are ICs, and physiological evidence reinforced the concept that ICs are involved in generation of rhythmicity and translation of neural inputs in gastrointestinal smooth muscles. Controlling the development of ICs provides a powerful new tool for the investigation of the physiological role of these cells.


Subject(s)
Digestive System/cytology , Gastrointestinal Motility/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Colony-Stimulating Factor/physiology , Animals , Animals, Newborn , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Colon/cytology , Colon/physiology , Digestive System/growth & development , Digestive System Physiological Phenomena , Ileum/cytology , Ileum/physiology , Methylene Blue , Mice , Mice, Inbred BALB C , Microscopy, Electron , Muscle, Smooth/chemistry , Muscle, Smooth/ultrastructure , Myenteric Plexus/chemistry , Myenteric Plexus/ultrastructure , Neural Conduction , Organelles/ultrastructure , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/immunology , Tolonium Chloride
18.
Leuk Lymphoma ; 11 Suppl 1: 131-7, 1993.
Article in English | MEDLINE | ID: mdl-7504543

ABSTRACT

Chronic myelogenous leukemia (CML) cell growth may be inhibited by exposure to antisense (AS) oligodeoxynucleotides (ODN). Our initial studies targeted the c-myb protooncogene and were carried out on cells derived from patients in CML blast crisis. Subsequently, we extended these studies to cells isolated from patients in chronic disease phase. We found that c-myb AS ODN inhibited growth of CML CFU-GM in a dose dependent, sequence specific manner in approximately 75% of cases evaluated. Bcr-abl expression was either greatly decreased or nondetectable in the residual colonies and no residual leukemic CFU were demonstrable upon re-plating of treated cells. AS ODN that target the c-kit protooncogene also inhibit CML CFU and lead to downregulation of bcr-abl in responding cells in approximately 50% of cases. Therefore, AS ODN may prove to be useful purging agents. Most recently, we have treated SCID mice engrafted with bcr-abl expressing human K562 cell leukemia with phosphorothioate modified AS ODN. We have found that treated mice survive three to eight times longer than their untreated or sense treated controls. In aggregate, these results suggest that AS ODN may prove useful for both ex vivo and in vivo treatment of patients with CML.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Neoplasm Proteins/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Colony-Stimulating Factor/antagonists & inhibitors , Animals , Base Sequence , Blast Crisis/pathology , Drug Screening Assays, Antitumor , Female , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Chronic-Phase/pathology , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Transplantation , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Proto-Oncogene Proteins c-myb , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Thionucleotides/therapeutic use , Tumor Cells, Cultured , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...