672 results on '"Receptor, Macrophage Colony-Stimulating Factor"'
Search Results
2. Discovery of BPR1R024, an Orally Active and Selective CSF1R Inhibitor that Exhibits Antitumor and Immunomodulatory Activity in a Murine Colon Tumor Model
- Author
-
Hsing Pang Hsieh, Pei-Chen Wang, Wan-Ching Yen, Kun-Hung Lee, Yu-Chen Huang, Chun-Yu Chang, Yu-Chieh Su, Chen-Ming Yang, You-Liang Lai, Wen-Hsing Lin, Chiung-Tong Chen, Chuan Shih, Teng-Kuang Yeh, Ling-Hui Chou, and Cai-Syuan Wu
- Subjects
Male ,medicine.medical_treatment ,Administration, Oral ,Antineoplastic Agents ,Receptor, Macrophage Colony-Stimulating Factor ,Rats, Sprague-Dawley ,Immunomodulating Agents ,Mice ,Structure-Activity Relationship ,Cancer immunotherapy ,Oral administration ,In vivo ,Cell Line, Tumor ,Drug Discovery ,medicine ,Animals ,Receptor ,Protein Kinase Inhibitors ,IC50 ,Tumor microenvironment ,Chemistry ,Kinase ,Neoplasms, Experimental ,Mice, Inbred C57BL ,Colonic Neoplasms ,Cancer research ,Molecular Medicine ,Aurora Kinase B - Abstract
Colony-stimulating factor-1 receptor (CSF1R) is implicated in tumor-associated macrophage (TAM) repolarization and has emerged as a promising target for cancer immunotherapy. Herein, we describe the discovery of orally active and selective CSF1R inhibitors by property-driven optimization of BPR1K871 (9), our clinical multitargeting kinase inhibitor. Molecular docking revealed an additional nonclassical hydrogen-bonding (NCHB) interaction between the unique 7-aminoquinazoline scaffold and the CSF1R hinge region, contributing to CSF1R potency enhancement. Structural studies of CSF1R and Aurora kinase B (AURB) demonstrated the differences in their back pockets, which inspired the use of a chain extension strategy to diminish the AURA/B activities. A lead compound BPR1R024 (12) exhibited potent CSF1R activity (IC50 = 0.53 nM) and specifically inhibited protumor M2-like macrophage survival with a minimal effect on antitumor M1-like macrophage growth. In vivo, oral administration of 12 mesylate delayed the MC38 murine colon tumor growth and reversed the immunosuppressive tumor microenvironment with the increased M1/M2 ratio.
- Published
- 2021
- Full Text
- View/download PDF
3. PTPRJ promotes osteoclast maturation and activity by inhibiting Cbl‐mediated ubiquitination of NFATc1 in late osteoclastogenesis
- Author
-
Moran Shalev, Sergey Kapishnikov, Esther Arman, Merle Stein, Isabelle Royal, Vlad Brumfeld, Ari Elson, Jan Tuckermann, and Yael Cohen-Sharir
- Subjects
Male ,0301 basic medicine ,Regulator ,Osteoclasts ,Receptor, Macrophage Colony-Stimulating Factor ,Protein tyrosine phosphatase ,Biochemistry ,Monocytes ,Bone resorption ,Osteoclast maturation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Multinucleate ,Ubiquitin ,Osteogenesis ,Osteoclast ,medicine ,Animals ,Proto-Oncogene Proteins c-cbl ,Molecular Biology ,Transcription factor ,Cells, Cultured ,NFATC Transcription Factors ,biology ,Chemistry ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,Ubiquitination ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Female - Abstract
Bone-resorbing osteoclasts (OCLs) are multinucleated phagocytes, whose central roles in regulating bone formation and homeostasis are critical for normal health and development. OCLs are produced from precursor monocytes in a multistage process that includes initial differentiation, cell-cell fusion, and subsequent functional and morphological maturation; the molecular regulation of osteoclastogenesis is not fully understood. Here, we identify the receptor-type protein tyrosine phosphatase PTPRJ as an essential regulator specifically of OCL maturation. Monocytes from PTPRJ-deficient (JKO) mice differentiate and fuse normally, but their maturation into functional OCLs and their ability to degrade bone are severely inhibited. In agreement, mice lacking PTPRJ throughout their bodies or only in OCLs exhibit increased bone mass due to reduced OCL-mediated bone resorption. We further show that PTPRJ promotes OCL maturation by dephosphorylating the M-CSF receptor (M-CSFR) and Cbl, thus reducing the ubiquitination and degradation of the key osteoclastogenic transcription factor NFATc1. Loss of PTPRJ increases ubiquitination of NFATc1 and reduces its amounts at later stages of osteoclastogenesis, thereby inhibiting OCL maturation. PTPRJ thus fulfills an essential and cell-autonomous role in promoting OCL maturation by balancing between the pro- and anti-osteoclastogenic activities of the M-CSFR and maintaining NFATc1 expression during late osteoclastogenesis.
- Published
- 2021
- Full Text
- View/download PDF
4. Conditional genetic deletion of CSF1 receptor in microglia ameliorates the physiopathology of Alzheimer’s disease
- Author
-
Vincent Pons, Pascal Lévesque, Serge Rivest, and Marie-Michèle Plante
- Subjects
0301 basic medicine ,Hippocampus ,Cognitive decline ,Plaque, Amyloid ,lcsh:RC346-429 ,Amyloid beta-Protein Precursor ,Mice ,0302 clinical medicine ,mCSF ,TREM2 ,Receptors, Immunologic ,Receptor ,Innate immunity ,Membrane Glycoproteins ,Microglia ,3. Good health ,medicine.anatomical_structure ,Neurology ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Amyloid ,β-Catenin ,Cognitive Neuroscience ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,lcsh:RC321-571 ,03 medical and health sciences ,Phagocytosis ,Alzheimer Disease ,medicine ,Dementia ,Animals ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,lcsh:Neurology. Diseases of the nervous system ,Innate immune system ,Amyloid beta-Peptides ,business.industry ,Research ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,nervous system ,IL-34 ,Immunology ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Background Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the most common form of dementia in the world. Microglia are the innate immune cells of CNS; their proliferation, activation, and survival in pathologic and healthy brain have previously been shown to be highly dependent on CSF1R. Methods Here, we investigate the impact of such receptor on AD etiology and microglia. We deleted CSF1R using Cre/Lox system; the knockout (KO) is restricted to microglia in the APP/PS1 mouse model. We induced the knockout at 3 months old, before plaque formation, and evaluated both 6- and 8-month-old groups of mice. Results Our findings demonstrated that CSF1R KO did not impair microglial survival and proliferation at 6 and 8 months of age in APP cKO compared to their littermate-control groups APPSwe/PS1. We have also shown that cognitive decline is delayed in CSF1R-deleted mice. Ameliorations of AD etiology are associated with a decrease in plaque volume in the cortex and hippocampus area. A compensating system seems to take place following the knockout, since TREM2/β-Catenin and IL-34 expression are significantly increased. Such a compensatory mechanism may promote microglial survival and phagocytosis of Aβ in the brain. Conclusions Our results provide new insights on the role of CSF1R in microglia and how it interacts with the TREM2/β-Catenin and IL-34 system to clear Aβ and ameliorates the physiopathology of AD.
- Published
- 2021
5. Blockade of the colony-stimulating factor-1 receptor reverses bone loss in osteoporosis mouse models
- Author
-
Rosa I. Acosta-González, Héctor Fabián Torres-Rodríguez, Juan Miguel Jimenez-Andrade, Arisai Martínez-Martínez, Enriqueta Muñoz-Islas, and Martha B. Ramírez-Rosas
- Subjects
medicine.medical_specialty ,Ovariectomy ,Osteoporosis ,Receptor, Macrophage Colony-Stimulating Factor ,Antibodies ,Streptozocin ,Diabetes Mellitus, Experimental ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Femur ,Femoral neck ,Pharmacology ,Mice, Inbred ICR ,business.industry ,Macrophage Colony-Stimulating Factor ,Osteopetrosis ,General Medicine ,Streptozotocin ,medicine.disease ,Blockade ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,medicine.anatomical_structure ,Endocrinology ,030220 oncology & carcinogenesis ,Female ,Cortical bone ,Secondary osteoporosis ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Mice lacking either colony-stimulating factor-1 (CSF-1) or its receptor, CSF-1R, display osteopetrosis. Accordingly, genetic deletion or pharmacological blockade of CSF-1 prevents the bone loss associated with estrogen deficiency. However, the role of CSF-1R in osteoporosis models of type-1 diabetes (T1D) and ovariectomy (OVX) has not been examined. Thus, we evaluated whether CSF-1R blockade would relieve the bone loss in a model of primary osteoporosis (female mice with OVX) and a model of secondary osteoporosis (female with T1D) using micro-computed tomography. Female ICR mice at 10 weeks underwent OVX or received five daily administrations of streptozotocin (ip, 50 mg/kg) to induce T1D. Four weeks after OVX and 14 weeks after first injection of streptozotocin, mice received an anti-CSF-1R (2G2) antibody (10 mg/kg, ip; once/week for 6 weeks) or vehicle. At the last day of antibody administration, mice were sacrificed and femur and tibia were harvested for micro-computed tomography analysis. Mice with OVX had a significant loss of trabecular bone at the distal femoral and proximal tibial metaphysis. Chronic treatment with anti-CSF-1R significantly reversed the trabecular bone loss at these anatomical sites. Streptozotocin-induced T1D resulted in significant loss of trabecular bone at the femoral neck and cortical bone at the femoral mid-diaphysis. Chronic treatment with anti-CSF-1R antibody significantly reversed the bone loss observed in mice with T1D. Our results demonstrate that blockade of CSF-1R signaling reverses bone loss in two different mouse models of osteoporosis.
- Published
- 2020
- Full Text
- View/download PDF
6. CSF-1 maintains pathogenic but not homeostatic myeloid cells in the central nervous system during autoimmune neuroinflammation
- Author
-
Daniel Hwang, Maryam S. Seyedsadr, Larissa Lumi Watanabe Ishikawa, Alexandra Boehm, Ziver Sahin, Giacomo Casella, Soohwa Jang, Michael V. Gonzalez, James P. Garifallou, Hakon Hakonarson, Weifeng Zhang, Dan Xiao, Abdolmohamad Rostami, Guang-Xian Zhang, and Bogoljub Ciric
- Subjects
Central Nervous System ,Macrophage colony-stimulating factor ,Encephalomyelitis, Autoimmune, Experimental ,Multiple Sclerosis ,Immunology ,Central nervous system ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,Mice ,Immune system ,Downregulation and upregulation ,medicine ,Animals ,Immunology and Allergy ,Myeloid Cells ,Benzothiazoles ,Picolinic Acids ,Neuroinflammation ,Multidisciplinary ,Microglia ,Macrophage Colony-Stimulating Factor ,Multiple sclerosis ,Experimental autoimmune encephalomyelitis ,medicine.disease ,Mice, Inbred C57BL ,medicine.anatomical_structure - Abstract
The receptor for colony stimulating factor 1 (CSF-1R) is important for the survival and function of myeloid cells that mediate pathology during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). CSF-1 and IL-34, the ligands of CSF-1R, have similar bioactivities but distinct tissue and context-dependent expression patterns, suggesting that they have different roles. This could be the case in EAE, given that CSF-1 expression is upregulated in the CNS, while IL-34 remains constitutively expressed. We found that targeting CSF-1 with neutralizing mAb halted ongoing EAE, with efficacy superior to CSF-1R inhibitor BLZ945, whereas IL-34 neutralization had no effect, suggesting that pathogenic myeloid cells were maintained by CSF-1, not IL-34. Both anti-CSF-1- and BLZ945-treatment greatly reduced numbers of monocyte-derived cells and microglia in the CNS. However, anti-CSF-1 selectively depleted inflammatory microglia and monocytes in inflamed CNS areas, whereas BLZ945 depleted virtually all myeloid cells, including quiescent microglia, throughout the CNS. Anti-CSF-1 treatments reduced the size of demyelinated lesions, and microglial activation in the grey matter. Lastly, we found that bone marrow-derived immune cells were the major mediators of CSF-1R-dependent pathology, while microglia played a lesser role. Our findings suggest that targeting CSF-1 could be effective in ameliorating MS pathology, while preserving the homeostatic functions of myeloid cells, thereby minimizing risks associated with total ablation of CSF-1R-dependent cells.Significance StatementMultiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), are autoimmune diseases characterized by accumulation of myeloid immune cells into the central nervous system (CNS). Both harmful and beneficial myeloid cells are present in EAE/MS, and a goal of MS therapy is to preferentially remove harmful myeloid cells. The receptor for CSF-1 (CSF-1R) is found on myeloid cells and it is important for their survival. CSF-1R can bind two ligands, CSF-1 and IL-34, but is unknown whether their functions in EAE/MS differ. We found that blocking CSF-1 depleted only harmful myeloid cells in the CNS and suppressed EAE, whereas blocking IL-34 had no effect. Thus, we propose that blocking CSF-1 could be a novel therapy for MS.
- Published
- 2022
- Full Text
- View/download PDF
7. CSF1/CSF1R signaling mediates malignant pleural effusion formation
- Author
-
Chrysavgi N. Kosti, Photene C. Vaitsi, Apostolos G. Pappas, Marianthi P. Iliopoulou, Katherina K. Psarra, Sophia F. Magkouta, and Ioannis T. Kalomenidis
- Subjects
Mice ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Macrophage Colony-Stimulating Factor ,Macrophages ,Colonic Neoplasms ,Animals ,Receptor Protein-Tyrosine Kinases ,Receptor, Macrophage Colony-Stimulating Factor ,General Medicine ,Pleural Effusion, Malignant - Abstract
Malignant pleural effusion (MPE) is an incurable common manifestation of many malignancies. Its formation is orchestrated by complex interactions among tumor cells, inflammatory cells, and the vasculature. Tumor-associated macrophages present the dominant inflammatory population of MPE, and M2 macrophage numbers account for dismal prognosis. M2 polarization is known to be triggered by CSF1/CSF1 receptor (CSF1R) signaling. We hypothesized that CSF1R+ M2 macrophages favor MPE formation and could be therapeutically targeted to limit MPE. We generated mice with CSF1R-deficient macrophages and induced lung and colon adenocarcinoma-associated MPE. We also examined the therapeutic potential of a clinically relevant CSF1R inhibitor (BLZ945) in lung and colon adenocarcinoma-induced experimental MPE. We showed that CSF1R+ macrophages promoted pleural fluid accumulation by enhancing vascular permeability, destabilizing tumor vessels, and favoring immune suppression. We also showed that CSF1R inhibition limited MPE in vivo by reducing vascular permeability and neoangiogenesis and impeding tumor progression. This was because apart from macrophages, CSF1R signals in cancer-associated fibroblasts leading to macrophage inflammatory protein 2 secretion triggered the manifestation of suppressive and angiogenic properties in macrophages upon CXCR2 paracrine activation. Pharmacological targeting of the CSF1/CSF1R axis can therefore be a vital strategy for limiting MPE.
- Published
- 2022
- Full Text
- View/download PDF
8. Macrophage-Targeting by CSF1/1R Blockade in Pancreatic Cancers
- Author
-
Won Jin Ho and Elizabeth M. Jaffee
- Subjects
Cancer Research ,T-Lymphocytes ,Receptor, Macrophage Colony-Stimulating Factor ,Receptors, Cell Surface ,Adenocarcinoma ,Deoxycytidine ,Article ,Cohort Studies ,Mice ,Random Allocation ,Tumor Microenvironment ,Animals ,Humans ,Lectins, C-Type ,Macrophage Colony-Stimulating Factor ,Macrophages ,Gemcitabine ,Mice, Inbred C57BL ,Pancreatic Neoplasms ,Mannose-Binding Lectins ,Oncology ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Tissue Array Analysis ,Female ,Immunotherapy ,Mannose Receptor ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Cancer immunotherapy generally offers limited clinical benefit without coordinated strategies to mitigate the immunosuppressive nature of the tumor microenvironment. Critical drivers of immune escape in the tumor microenvironment include tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC), which not only mediate immune suppression but also promote metastatic dissemination and impart resistance to cytotoxic therapies. Thus, strategies to ablate the effects of these myeloid cell populations may offer great therapeutic potential. In this report, we demonstrate in a mouse model of pancreatic ductal adenocarcinoma (PDAC) that inhibiting signaling by the myeloid growth factor receptor CSF1R can functionally reprogram macrophage responses that enhance antigen presentation and productive anti-tumor T cell responses. Investigations of this response revealed that CSF1R blockade also upregulated T cell checkpoint molecules, including PDL1 and CTLA4, thereby restraining beneficial therapeutic effects. We found that PD1 and CTLA4 antagonists showed limited efficacy as single agents to restrain PDAC growth, but that that combining these agents with CSF1R blockade potently elicited tumor regressions, even in larger established tumors. Taken together, our findings provide a rationale to reprogram immunosuppressive myeloid cell populations in the tumor microenvironment under conditions that can significantly empower the therapeutic effects of checkpoint-based immunotherapeutics.
- Published
- 2021
9. Resident Macrophages in the Heart: Cardioprotective Under Pressure
- Author
-
Sumanth D. Prabhu and Mohamed Ameen Ismahil
- Subjects
medicine.medical_specialty ,Physiology ,Receptors, CCR2 ,heart failure ,Neovascularization, Physiologic ,Cardiomegaly ,Receptor, Macrophage Colony-Stimulating Factor ,Article ,Mice ,Fibrosis ,Internal medicine ,medicine ,Animals ,Ventricular remodeling ,Cells, Cultured ,Original Research ,business.industry ,Macrophages ,Myocardium ,Heart ,medicine.disease ,Mice, Inbred C57BL ,Heart failure ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Cardiology ,Single-Cell Analysis ,hypertrophy ,monocytes ,Cardiology and Cardiovascular Medicine ,business ,Transcriptome - Abstract
Supplemental Digital Content is available in the text., Rationale: The initial hypertrophy response to cardiac pressure overload is considered compensatory, but with sustained stress, it eventually leads to heart failure. Recently, a role for recruited macrophages in determining the transition from compensated to decompensated hypertrophy has been established. However, whether cardiac resident immune cells influence the early phase of hypertrophy development has not been established. Objective: To assess the role of cardiac immune cells in the early hypertrophy response to cardiac pressure overload induced by transverse aortic constriction (TAC). Methods and Results: We performed cytometry by time-of-flight to determine the identity and abundance of immune cells in the heart at 1 and 4 weeks after TAC. We observed a substantial increase in cardiac macrophages 1 week after TAC. We then conducted Cite-Seq single-cell RNA sequencing of cardiac immune cells isolated from 4 sham and 6 TAC hearts. We identified 12 clusters of monocytes and macrophages, categorized as either resident or recruited macrophages, that showed remarkable changes in their abundance between sham and TAC conditions. To determine the role of cardiac resident macrophages early in the response to a hypertrophic stimulus, we used a blocking antibody against macrophage colony-stimulating factor 1 receptor (CD115). As blocking CD115 initially depletes all macrophages, we allowed the replenishment of recruited macrophages by monocytes before performing TAC. This preferential depletion of resident macrophages resulted in enhanced fibrosis and a blunted angiogenesis response to TAC. Macrophage depletion in CCR2 (C-C chemokine receptor type 2) knockout mice showed that aggravated fibrosis was primarily caused by the recruitment of monocyte-derived macrophages. Finally, 6 weeks after TAC these early events lead to depressed cardiac function and enhanced fibrosis, despite complete restoration of cardiac immune cells. Conclusions: Cardiac resident macrophages are a heterogeneous population of immune cells with key roles in stimulating angiogenesis and inhibiting fibrosis in response to cardiac pressure overload.
- Published
- 2021
10. Discovery of (
- Author
-
Qi, Lv, Xiang, Pan, Dan, Wang, Quanjin, Rong, Ben, Ma, Xiaolong, Xie, Yinan, Zhang, Junwei, Wang, and Lihong, Hu
- Subjects
Mice, Inbred BALB C ,Chemotaxis ,Macrophages ,Administration, Oral ,Mice, Nude ,Antineoplastic Agents ,Receptor, Macrophage Colony-Stimulating Factor ,Molecular Docking Simulation ,Mice ,Structure-Activity Relationship ,Cell Line, Tumor ,Drug Discovery ,Animals ,Humans ,Urea ,Female ,Immunotherapy ,Colorectal Neoplasms - Abstract
Inhibiting the polarization or survival of tumor-associated macrophages through blocking CSF-1/CSF-1R signal transduction has become a promising strategy for cancer immunotherapy. Herein, a series of (
- Published
- 2021
11. IL-34 reprograms glycolytic and osteoclastic RA macrophages via Syndecan-1 and M-CSFR
- Author
-
Van Raemdonck, Katrien, Umar, Sadiq, Palasiewicz, Karol, Volin, Michael V., Elshabrawy, Hatem A., Romay, Bianca, Tetali, Chandana, Ahmed, Azam, Amin, M. Asif, Zomorrodi, Ryan K., Sweiss, Nadera, and Shahrara, Shiva
- Subjects
Arthritis, Rheumatoid ,Inflammation ,Mice ,Interleukins ,Macrophages ,Synovial Membrane ,Animals ,Osteoclasts ,Receptor, Macrophage Colony-Stimulating Factor ,Syndecan-1 ,Phosphorylation ,Glycolysis ,Article - Abstract
OBJECTIVE: In RA, IL-34 serum levels are linked with increased disease severity. IL-34 binds to two receptors, M-CSFR and Syndecan-1 (SDC-1), which are co-expressed in RA macrophages (MФs). Expression of both IL-34 and SDC-1 is strikingly elevated in the RA synovium, yet their mechanism of action remains undefined. METHODS: To characterize the significance of IL-34 in immunometabolism, its mechanism of action was elucidated in joint MФs, fibroblasts and effector T cells using RA and preclinical models. RESULTS: Intriguingly, SDC-1 activates IL-34-induced M-CSFR phosphorylation and reprograms RA naïve cells into distinctive CD14(+)CD86(+)GLUT1(+)M34 MФs that express elevated levels of IL-1β, CXCL8 and CCL2. In murine M34 MФs, the inflammatory phenotype is accompanied by potentiated glycolytic activity, exhibited by transcriptional upregulation of GLUT1, C-MYC and HIF1α and amplified pyruvate and L-lactate secretion. Local expression of IL-34 provokes arthritis, by expanding the glycolytic F4/80(+)iNOS(+)MΦ population, which in turn attracts fibroblasts and polarizes Th1/Th17 cells. The crosstalk between murine M34 MΦs and Th1/Th17 cells broadens the inflammatory and metabolic phenotypes resulting in the expansion of IL-34 pathogenicity. Consequently, IL-34-instigated joint inflammation was alleviated in RAG−/− compared to wild type mice. Consistently, SDC-1 deficiency attenuated IL-34-induced arthritis by interfering with joint glycolytic M34 MΦ and osteoclast remodeling. Similarly, inhibition of glycolysis by 2-DG reversed the joint swelling and metabolic rewiring triggered by IL-34 via HIF1α and C-MYC induction. CONCLUSION: IL-34 is a novel endogenous factor that remodels hypermetabolic M34 MΦs and facilitates their cross-regulation with effector T cells to advance RA inflammatory bone destruction.
- Published
- 2021
12. Alleviation of neuropathic pain by over-expressing a soluble colony-stimulating factor 1 receptor to suppress microgliosis and macrophage accumulation
- Author
-
Haripriya Sivakumar, Xuenong Bo, Ping K. Yip, Jie Li, Glesni A. Parry, Min Liu, and Svetlana Gushchina
- Subjects
medicine.medical_specialty ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,Microgliosis ,Colony stimulating factor 1 receptor ,Cellular and Molecular Neuroscience ,Mice ,Lumbar ,Internal medicine ,Ganglia, Spinal ,medicine ,Animals ,Receptor ,Microglia ,Macrophage Colony-Stimulating Factor ,Macrophages ,Spinal cord ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,Neurology ,Spinal Cord ,Hyperalgesia ,Neuropathic pain ,Neuralgia ,Sciatic nerve - Abstract
Microglial proliferation and activation and macrophage accumulation are implicated in neuropathic pain development. In this study, we aim to suppress microgliosis and macrophage accumulation by over-expressing a non-functional soluble colony stimulating factor-1 receptor (sCSF1R) using an adeno-associated virus 9 vector (AAV9). AAV9/sCSF1R and the control vector AAV9/GFP were intrathecally administered into the lumbar spine of adult C57BL/6 mice. Two weeks later, these mice underwent partial sciatic nerve ligation to induce neuropathic pain. GFP and sCSF1R were highly expressed in lumbar dorsal root ganglia (DRG) and spinal cord of AAV9-injected mice. A significant increase in microglia densities in the dorsal and ventral horns of lumbar spinal cords and macrophage densities in DRG and sciatic nerves were observed in the mice with either ligation alone or pre-treated with AAV9/GFP. In nerve-ligated mice pre-treated with AAV9/sCSF1R the microglia densities in the dorsal and ventral horns and macrophage densities in DRG and sciatic nerves were significantly lower compared to nerve-ligated mice pre-treated with AAV9/GFP. Behavioral tests showed that nerve-ligated mice pre-treated with AAV9/sCSF1R had a significantly higher paw withdrawal threshold, indicating the alleviation of neuropathic pain. The results implicate that viral vector-mediated expression of sCSF1R may represent a novel strategy in the alleviation of neuropathic pain.
- Published
- 2021
13. M-CSF Receptor Antagonists Inhibit the Initiation and Progression of Hepatocellular Carcinoma in Mice
- Author
-
Hiroshi Kono, Michio Hara, Shinji Furuya, Chao Sun, Hisataka Fukushima, Daisuke Ichikawa, Yoshihiro Akazawa, Yuuki Nakata, Hiroyuki Wakana, and Hideki Fujii
- Subjects
Male ,Cancer Research ,Carcinoma, Hepatocellular ,Kupffer Cells ,Angiogenesis ,medicine.drug_class ,medicine.medical_treatment ,Receptor, Macrophage Colony-Stimulating Factor ,Anisoles ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Hepatic carcinogenesis ,medicine ,Animals ,Macrophage ,Receptor ,Saline ,Cell Proliferation ,Treated group ,business.industry ,Macrophages ,Liver Neoplasms ,General Medicine ,medicine.disease ,Receptor antagonist ,Immunohistochemistry ,Tumor Burden ,Cell Transformation, Neoplastic ,Pyrimidines ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Cancer research ,business ,Biomarkers - Abstract
Background/aim The aim of this study was to investigate the effects of the macrophage colony-stimulating factor (M-CSF) receptor antagonist on hepatic carcinogenesis in mice. Materials and methods Mice were injected with diethylnitrosamine (DEN) and treated with M-CSF receptor antagonist GW2580 (GW) or a saline vehicle just after (early treated group) or 2 weeks after (late treated group) DEN injection. Animals were sacrificed after 28 weeks and incidence of tumor was assessed. Isolated Kupffer cells were co-cultured with M-CSF in the presence or absence of GW, and the concentration of VEGF was measured. Results The incidence of tumors was significantly blunted both in the early- and the late-treated groups. In addition, angiogenesis within the tumor was also suppressed in both groups. The concentration of VEGF increased in Kupffer cells treated with M-CSF compared to those cultured without M-CSF. This increase was blunted by GW. Conclusion M-CSF and its receptor could be novel molecular targets for hepatocellular carcinoma.
- Published
- 2019
- Full Text
- View/download PDF
14. Preclinical evaluation of 3D185, a novel potent inhibitor of FGFR1/2/3 and CSF-1R, in FGFR-dependent and macrophage-dominant cancer models
- Author
-
Yanyan Shen, Yang Dai, Yi Su, Yi Chen, Yueliang Wang, Bo Liu, Yuchen Jiang, Zuoquan Xie, Yinchun Ji, Chuantao Zha, Deqiao Sun, Pengcong Hou, Xia Peng, Meiyu Geng, Jian Ding, and Jing Ai
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,T cell ,Mice, Nude ,Apoptosis ,Receptor, Macrophage Colony-Stimulating Factor ,lcsh:RC254-282 ,Piperazines ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,In vivo ,Neoplasms ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Animals ,Humans ,Receptor, Fibroblast Growth Factor, Type 3 ,Receptor, Fibroblast Growth Factor, Type 1 ,Kinase activity ,Phosphorylation ,Receptor, Fibroblast Growth Factor, Type 2 ,Protein Kinase Inhibitors ,Cell Proliferation ,Tumor microenvironment ,Mice, Inbred BALB C ,Kinase inhibitor ,Chemistry ,FGFR ,Research ,Macrophages ,3D185 ,Cell migration ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Signal Transduction ,CSF-1R - Abstract
Background The interaction between tumor cells and their immunosuppressive microenvironment promotes tumor progression and drug resistance. Thus, simultaneously targeting tumor cells and stromal cells is expected to have synergistic antitumor effects. Herein, we present for the first time a preclinical antitumor investigation of 3D185, a novel dual inhibitor targeting FGFRs, which are oncogenic drivers, and CSF-1R, which is the major survival factor for protumor macrophages. Methods The antitumor characteristics of 3D185 were assessed by a range of assays, including kinase profiling, cell viability, cell migration, immunoblotting, CD8+ T cell suppression, and in vivo antitumor efficacy, followed by flow cytometric and immunohistochemical analyses of tumor-infiltrating immune cells and endothelial cells in nude mice and immune-competent mice. Results 3D185 significantly inhibited the kinase activity of FGFR1/2/3 and CSF-1R, with equal potency and high selectivity over other kinases. 3D185 suppressed FGFR signaling and tumor cell growth in FGFR-driven models both in vitro and in vivo. In addition, 3D185 could inhibit the survival and M2-like polarization of macrophages, reversing the immunosuppressive effect of macrophages on CD8+ T cells as well as CSF1-differentiated macrophage induced-FGFR3-aberrant cancer cell migration. Furthermore, 3D185 inhibited tumor growth via remodeling the tumor microenvironment in TAM-dominated tumor models. Conclusions 3D185 is a promising antitumor candidate drug that simultaneously targets tumor cells and their immunosuppressive microenvironment and has therapeutic potential due to synergistic effects. Our study provides a solid foundation for the investigation of 3D185 in cancer patients, particularly in patients with aberrant FGFR and abundant macrophages, who respond poorly to classic pan-FGFRi treatment. Electronic supplementary material The online version of this article (10.1186/s13046-019-1357-y) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
15. CSF-1 controls cerebellar microglia and is required for motor function and social interaction
- Author
-
Violeta Chitu, Samuel A. Rose, Anne Schaefer, Aleksandra Wroblewska, Brian D. Brown, Ana Badimon, Lotje de Witte, Zhenyu Yue, Kazuhiko Yamamuro, Maria Casanova-Acebes, Andrew Leader, Pinar Ayata, Alessia Baccarini, Florent Ginhoux, I-li Tan, Yonit Lavin, Hortense Le Bourhis, Scott J. Russo, Veronika Kana, Christie Chang, Alexandra L. Joyner, Eric S. Sweet, Peter See, Hirofumi Morishita, Navpreet Tung, Miriam Merad, Elisa M. Nabel, E. Richard Stanley, Fiona Desland, Meghan E. Flanigan, and Marjolein A. M. Sneeboer
- Subjects
0301 basic medicine ,Macrophage Colony-Stimulating Factor ,Immunology ,Science program ,Library science ,Receptor, Macrophage Colony-Stimulating Factor ,Motor function ,Article ,03 medical and health sciences ,Mice ,030104 developmental biology ,0302 clinical medicine ,Innovator ,Political science ,Immunology and Allergy ,Animals ,Interpersonal Relations ,Microglia ,10. No inequality ,030217 neurology & neurosurgery ,Research Articles ,Signal Transduction - Abstract
Microglia are a heterogeneous population whose identity and function are dictated by signals from their microenvironment. Kana et al. show CSF-1 signaling is critical for cerebellar microglial transcriptional identity and homeostasis, and that altering the CSF-1–CSF-1R axis leads to motor and behavioral defects., Microglia, the brain resident macrophages, critically shape forebrain neuronal circuits. However, their precise function in the cerebellum is unknown. Here we show that human and mouse cerebellar microglia express a unique molecular program distinct from forebrain microglia. Cerebellar microglial identity was driven by the CSF-1R ligand CSF-1, independently of the alternate CSF-1R ligand, IL-34. Accordingly, CSF-1 depletion from Nestin+ cells led to severe depletion and transcriptional alterations of cerebellar microglia, while microglia in the forebrain remained intact. Strikingly, CSF-1 deficiency and alteration of cerebellar microglia were associated with reduced Purkinje cells, altered neuronal function, and defects in motor learning and social novelty interactions. These findings reveal a novel CSF-1–CSF-1R signaling-mediated mechanism that contributes to motor function and social behavior.
- Published
- 2019
16. Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response
- Author
-
Carola Ries, Chia Huey Ooi, Kevin Kos, Seth B. Coffelt, Antoinette van Weverwijk, Kelly Kersten, Ji-Ying Song, Joachim L. Schultze, Jos Jonkers, Philippe A. Cassier, Camilla Salvagno, Karin E. de Visser, Sander Tuit, Metamia Ciampricotti, Dominik Rüttinger, Kim Vrijland, Thomas Ulas, and Cheei-Sing Hau
- Subjects
medicine.medical_treatment ,secondary [Mammary Neoplasms, Experimental] ,Mice ,0302 clinical medicine ,Interferon ,Antineoplastic Combined Chemotherapy Protocols ,Mice, Knockout ,drug effects [Macrophages] ,0303 health sciences ,physiology [Interferon Type I] ,Antibodies, Monoclonal ,Immunosuppression ,3. Good health ,Cell biology ,therapeutic use [Antineoplastic Combined Chemotherapy Protocols] ,030220 oncology & carcinogenesis ,Interferon Type I ,Female ,therapeutic use [Antibodies, Monoclonal] ,medicine.drug ,drug effects [Immunity, Innate] ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Antibodies, Monoclonal, Humanized ,Article ,03 medical and health sciences ,Breast cancer ,therapeutic use [Cisplatin] ,Immunity ,ddc:570 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,030304 developmental biology ,Cisplatin ,pathology [Mammary Neoplasms, Experimental] ,Chemotherapy ,drug therapy [Mammary Neoplasms, Experimental] ,business.industry ,Macrophages ,immunology [Mammary Neoplasms, Experimental] ,Mammary Neoplasms, Experimental ,Cancer ,Cell Biology ,medicine.disease ,Immunity, Innate ,Blockade ,Cancer research ,antagonists & inhibitors [Receptor, Macrophage Colony-Stimulating Factor] ,business ,emactuzumab - Abstract
Recent studies have revealed a role for macrophages and neutrophils in limiting chemotherapy efficacy; however, the mechanisms underlying the therapeutic benefit of myeloid-targeting agents in combination with chemotherapy are incompletely understood. Here, we show that targeting tumour-associated macrophages by colony-stimulating factor-1 receptor (CSF-1R) blockade in the K14cre;Cdh1F/F;Trp53F/F transgenic mouse model for breast cancer stimulates intratumoural type I interferon (IFN) signalling, which enhances the anticancer efficacy of platinum-based chemotherapeutics. Notably, anti-CSF-1R treatment also increased intratumoural expression of type I IFN-stimulated genes in patients with cancer, confirming that CSF-1R blockade is a powerful strategy to trigger an intratumoural type I IFN response. By inducing an inflamed, type I IFN-enriched tumour microenvironment and by further targeting immunosuppressive neutrophils during cisplatin therapy, antitumour immunity was activated in this poorly immunogenic breast cancer mouse model. These data illustrate the importance of breaching multiple layers of immunosuppression during cytotoxic therapy to successfully engage antitumour immunity in breast cancer.
- Published
- 2019
- Full Text
- View/download PDF
17. Inhibition of TAMs improves the response to docetaxel in castration-resistant prostate cancer
- Author
-
Ping Tan, Wei Guan, Hua Xu, Zhuang Tang, Gideon Bollag, Junhui Hu, Lu Yang, Lily Wu, and Brian L. West
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Treatment response ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Aminopyridines ,Antineoplastic Agents ,Receptor, Macrophage Colony-Stimulating Factor ,Docetaxel ,Castration resistant ,urologic and male genital diseases ,Article ,Cell Line ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Endocrinology ,medicine ,Animals ,Humans ,Pyrroles ,Tumor growth ,Receptor ,neoplasms ,Chemotherapy ,business.industry ,Macrophages ,Antagonist ,medicine.disease ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Cytokines ,business ,medicine.drug - Abstract
For men with castration-resistant prostate cancer (CRPC), androgen-deprivation therapy (ADT) often becomes ineffective requiring the addition of docetaxel, a proven effective chemotherapy option. Tumor-associated macrophages (TAMs) are known to provide protumorigenic influences that contribute to treatment failure. In this study, we examined the contribution of TAMs to docetaxel treatment. An increased infiltration of macrophages in CRPC tumors was observed after treatment with docetaxel. Prostate cancer cells treated with docetaxel released more macrophage colony-stimulating factor (M-CSF-1 or CSF-1), IL-10 and other factors, which can recruit and modulate circulating monocytes to promote their protumorigenic functions. Inhibition of CSF-1 receptor kinase signaling with a small molecule antagonist (PLX3397) in CRPC models significantly reduces the infiltration of TAMs and their influences. As such, the addition of PLX3397 to docetaxel treatment resulted in a more durable tumor growth suppression than docetaxel alone. This study reveals a rational strategy to abrogate the influences of TAMs and extend the treatment response to docetaxel in CRPC.
- Published
- 2019
- Full Text
- View/download PDF
18. Modeling CSF-1 receptor deficiency diseases - how close are we?
- Author
-
Solen Gokhan, E. Richard Stanley, and Violeta Chitu
- Subjects
Microglia ,business.industry ,Neurodegeneration ,Leukodystrophy ,Receptor Protein-Tyrosine Kinases ,Receptor, Macrophage Colony-Stimulating Factor ,Cell Biology ,medicine.disease ,Biochemistry ,Penetrance ,Dysosteosclerosis ,Leukoencephalopathy ,Mice ,medicine.anatomical_structure ,Leukoencephalopathies ,Immunology ,Mutation ,medicine ,Macrophage ,Animals ,Humans ,Receptor ,business ,Molecular Biology ,Osteosclerosis - Abstract
The role of colony stimulating factor-1 receptor (CSF-1R) in macrophage and organismal development has been extensively studied in mouse. Within the last decade mutations in the CSF1R have been shown to cause rare diseases of both pediatric (Brain Abnormalities, Neurodegeneration, and Dysosteosclerosis (BANDDOS), OMIM #618476) and adult [CSF1R-related leukoencephalopathy (CRL), OMIM #221820] onset. Here we review the genetics, penetrance and histopathological features of these diseases and discuss to what extent the animal models of Csf1r deficiency currently available provide systems in which to study the underlying mechanisms involved.
- Published
- 2021
19. Aging and CNS Myeloid Cell Depletion Attenuate Breast Cancer Brain Metastasis
- Author
-
Lalage M. Wakefield, Patricia S. Steeg, Debbie Wei, Alex Man Lai Wu, Maxwell P. Lee, Dorian B. McGavern, Imran Khan, Andy D. Tran, Kristine A. Isanogle, Monika A. Chung, Quentin R. Smith, Christina Robinson, Selamawit Gossa, Ramakrishna Samala, Emma L. Dolan, Simone Difilippantonio, Yu-an Yang, Howard H. Yang, Helen R. Thorsheim, Brunilde Gril, and Esra Taner
- Subjects
Central Nervous System ,Cancer Research ,Myeloid ,Cell ,Receptor, Macrophage Colony-Stimulating Factor ,Triple Negative Breast Neoplasms ,Article ,Mice ,Immune system ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Myeloid Cells ,Microglia ,biology ,business.industry ,Brain Neoplasms ,Age Factors ,medicine.disease ,Astrogliosis ,medicine.anatomical_structure ,Oncology ,Cancer research ,biology.protein ,Antibody ,business ,Brain metastasis - Abstract
Purpose: Breast cancer diagnosed in young patients is often aggressive. Because primary breast tumors from young and older patients have similar mutational patterns, we hypothesized that the young host microenvironment promotes more aggressive metastatic disease. Experimental Design: Triple-negative or luminal B breast cancer cell lines were injected into young and older mice side-by-side to quantify lung, liver, and brain metastases. Young and older mouse brains, metastatic and naïve, were analyzed by flow cytometry. Immune populations were depleted using antibodies or a colony-stimulating factor-1 receptor (CSF-1R) inhibitor, and brain metastasis assays were conducted. Effects on myeloid populations, astrogliosis, and the neuroinflammatory response were determined. Results: Brain metastases were 2- to 4-fold higher in young as compared with older mouse hosts in four models of triple-negative or luminal B breast cancer; no age effect was observed on liver or lung metastases. Aged brains, naïve or metastatic, contained fewer resident CNS myeloid cells. Use of a CSF-1R inhibitor to deplete myeloid cells, including both microglia and infiltrating macrophages, preferentially reduced brain metastasis burden in young mice. Downstream effects of CSF-1R inhibition in young mice resembled that of an aged brain in terms of myeloid numbers, induction of astrogliosis, and Semaphorin 3A secretion within the neuroinflammatory response. Conclusions: Host microenvironmental factors contribute to the aggressiveness of triple-negative and luminal B breast cancer brain metastasis. CSF-1R inhibitors may hold promise for young brain metastasis patients.
- Published
- 2021
20. The receptor of the colony-stimulating factor-1 (CSF-1R) is a novel prognostic factor and therapeutic target in follicular lymphoma
- Author
-
Maria C. Cid, Dolors Colomer, Silvia Martín, Alba Matas-Céspedes, Martina Guerrero-Hernández, Joaquim Carreras, Anella Yahiaoui, Laura Magnano, Patricia Pérez-Galán, Alfredo Rivas-Delgado, Elias Campo, Neus Serrat, Lluis Hernández, Fabian Arenas, Juan G. Valero, Olga Balagué, Vanina Rodriguez, Armando López-Guillermo, Cristina Capdevila, and Stacey Tannheimer
- Subjects
Cancer microenvironment ,Macrophage colony-stimulating factor ,Cancer Research ,Angiogenesis ,Follicular lymphoma ,Aminopyridines ,Apoptosis ,Receptor, Macrophage Colony-Stimulating Factor ,Monocytes ,Article ,Mice ,Limfoma fol·licular ,In vivo ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Pyrroles ,Phosphorylation ,Lymphoma, Follicular ,Cell Proliferation ,Follicular dendritic cells ,B-cell lymphoma ,business.industry ,Macrophages ,Monocyte ,Germinal center ,Cell Differentiation ,Hematology ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Cancer research ,Càncer -- Tractament ,business ,Ex vivo - Abstract
Microenvironment contributes to follicular lymphoma (FL) pathogenesis and impacts survival with macrophages playing a controversial role. In the present study, using FL primary samples and HK follicular dendritic cells (FDC) to mimic the germinal center, together with mouse models, we have analyzed the three-way crosstalk of FL-FDC-macrophages and derived therapeutic opportunities. Ex vivo primary FL-FDC co-cultures (n = 19) and in vivo mouse co-xenografts demonstrated that FL-FDC crosstalk favors tumor growth and, via the secretion of CCL2 and CSF-1, promotes monocyte recruitment, differentiation, and polarization towards an M2-like protumoral phenotype. Moreover, FL-M2 co-cultures displayed enhanced angiogenesis, dissemination, and immunosuppression. Analysis of the CSF-1/CSF-1R pathway uncovered that CSF-1 was significantly higher in serum from grade 3A FL patients, and that high CSF-1R expression in FL biopsies correlated with grade 3A, reduced overall survival and risk of transformation. Furthermore, CSF-1R inhibition with pexidartinib (PLX3397) preferentially affected M2-macrophage viability and polarization program disrupting FL-M2 positive crosstalk. In vivo CSF1-R inhibition caused M2 reduction and repolarization towards M1 macrophages and antitumor effect cooperating with anti-CD20 rituximab. In summary, these results support the role of macrophages in FL pathogenesis and indicate that CSF-1R may be a relevant prognostic factor and a novel therapeutic target cooperating with anti-CD20 immunotherapy. Grants that contributed to this work included: Gilead Sciences Research Funded Agreement, Spanish Ministry of Economy and Competitiveness & European Regional Development Fund (ERDF) “Una manera de hacer Europa” for SAF2014/57708R and SAF2017/ 88275R to PP-G and MCC, RTI2018-094584-B-I00 to DC, CIBERONC (CB16/12/00334 and CB16/12/00225), and finally Generalitat de Catalunya support for AGAUR 2017SGR1009 to DC
- Published
- 2021
21. The Inhibitory Role of Rab11b in Osteoclastogenesis Through Triggering Lysosome-Induced Degradation of c-Fms and RANK Surface Receptors
- Author
-
Keiji Naruse, Yunxia Feng, Chiharu Sogawa, Masatoshi Morimatsu, Kuniaki Okamoto, Tomoko Kadowaki, Penggong Wei, Takayuki Tsukuba, Yuka Okusha, Eiko Sakai, Takanori Eguchi, Manh Tien Tran, and Hirohiko Okamura
- Subjects
Male ,0301 basic medicine ,Endocytic recycling ,RANK ,lcsh:Chemistry ,Mice ,0302 clinical medicine ,Osteogenesis ,vesicular transport ,Receptor ,lcsh:QH301-705.5 ,Cells, Cultured ,Spectroscopy ,Receptor Activator of Nuclear Factor-kappa B ,biology ,Chemistry ,Cell Differentiation ,General Medicine ,Computer Science Applications ,Cell biology ,medicine.anatomical_structure ,RANKL ,NFATc-1 ,030220 oncology & carcinogenesis ,symbols ,Endosome ,Receptor, Macrophage Colony-Stimulating Factor ,Article ,Catalysis ,Cell Line ,Inorganic Chemistry ,03 medical and health sciences ,symbols.namesake ,Osteoclast ,Lysosome ,medicine ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,NFATC Transcription Factors ,Endoplasmic reticulum ,Organic Chemistry ,Golgi apparatus ,c-Fms ,Mice, Inbred C57BL ,Rab11b ,030104 developmental biology ,osteoclasts ,lcsh:Biology (General) ,lcsh:QD1-999 ,rab GTP-Binding Proteins ,Proteolysis ,biology.protein ,Lysosomes - Abstract
Rab11b, abundantly enriched in endocytic recycling compartments, is required for the establishment of the machinery of vesicle trafficking. Yet, no report has so far characterized the biological function of Rab11b in osteoclastogenesis. Using in vitro model of osteoclasts differentiated from murine macrophages like RAW-D cells or bone marrow-derived macrophages, we elucidated that Rab11b served as an inhibitory regulator of osteoclast differentiation sequentially via (i) abolishing surface abundance of RANK and c-Fms receptors, and (ii) attenuating nuclear factor of activated T-cells c1 (NFATc-1) upstream signaling cascades, following RANKL stimulation. Rab11b was localized in early and late endosomes, Golgi complex, and endoplasmic reticulum, moreover, its overexpression enlarged early and late endosomes. Upon inhibition of lysosomal function by a specific blocker, chloroquine (CLQ), we comprehensively clarified a novel function of lysosomes on mediating proteolytic degradation of c-Fms and RANK surface receptors, drastically ameliorated by Rab11b overexpression in RAW-D cell-derived osteoclasts. These findings highlight the key role of Rab11b as an inhibitor of osteoclastogenesis by directing the transport of c-Fms and RANK surface receptors to lysosomes for degradation via the axis of early endosomes-late endosomes-lysosomes, thereby contributing towards the systemic equilibrium of the bone resorption phase.
- Published
- 2020
- Full Text
- View/download PDF
22. A transgenic line that reports CSF1R protein expression provides a definitive marker for the mouse mononuclear phagocyte system
- Author
-
Neil E. Humphreys, Martin D. Ryan, Clare Pridans, Antony Adamson, Anuj Sehgal, Veera A. Verkasalo, Derya D. Ozdemir, Garry A. Luke, Peter Hohenstein, Jennifer Stables, Katharine M. Irvine, Cheyenne J. Sandrock, Allison R. Pettit, Evi Wollscheid-Lengeling, Werner Mueller, David A. Hume, Rocio Rojo, and Kathleen Grabert
- Subjects
Myeloid ,Transgene ,Immunology ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,Monocytes ,Tendons ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Macrophage ,Animals ,RNA, Messenger ,Progenitor cell ,Muscle, Skeletal ,Mononuclear Phagocyte System ,Macrophage Colony-Stimulating Factor ,Macrophages ,Cell Differentiation ,Mononuclear phagocyte system ,Dendritic Cells ,Hematopoietic Stem Cells ,Cell biology ,Mice, Inbred C57BL ,Haematopoiesis ,medicine.anatomical_structure ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Genetics & genetic processes [F10] [Life sciences] ,Bone marrow ,Stem cell ,Génétique & processus génétiques [F10] [Sciences du vivant] ,Biomarkers ,030215 immunology - Abstract
The proliferation, differentiation and survival of cells of the mononuclear phagocyte system (MPS, progenitors, monocytes, macrophages and classical dendritic cells) is controlled by signals from the macrophage colony-stimulating factor receptor (CSF1R). Cells of the MPS lineage have been identified using numerous surface markers and transgenic reporters but none is both universal and lineage-restricted. Here we report the development and characterization of a novel CSF1R reporter mouse. A Fusion Red (FRed) cassette was inserted in-frame with the C-terminus of CSF1R, separated by a T2A-cleavable linker. The insertion had no effect of CSF1R expression or function. CSF1R-FRed was expressed in monocytes and macrophages and absent from granulocytes and lymphocytes. In bone marrow, CSF1R-FRed was absent in lineage-negative hematopoietic stem cells (HSC), arguing against a direct role for CSF1R in myeloid lineage commitment. It was highly-expressed in marrow monocytes and common myeloid progenitors (CMP) but significantly lower in granulocyte-macrophage progenitors (GMP). In sections of bone marrow, CSF1R-FRed was also detected in osteoclasts, CD169+ resident macrophages and, consistent with previous mRNA analysis, in megakaryocytes. In lymphoid tissues, CSF1R-FRed highlighted diverse MPS populations including classical dendritic cells. Whole mount imaging of non-lymphoid tissues in mice with combined CSF1R-FRed/Csf1r-EGFP confirmed the restriction of CSF1R expression to MPS cells. The two markers highlight the remarkable abundance and regular distribution of tissue MPS cells including novel macrophage populations within tendon and skeletal muscle and underlying the mesothelial/serosal/capsular surfaces of every major organ. The CSF1R-FRed mouse provides a novel reporter with exquisite specificity for cells of the MPS.
- Published
- 2020
- Full Text
- View/download PDF
23. Therapeutic Targeting of Retinal Immune Microenvironment With CSF-1 Receptor Antibody Promotes Visual Function Recovery After Ischemic Optic Neuropathy
- Author
-
Yizhen Tang, Zebin Xiao, Li Pan, Dongli Zhuang, Kin-Sang Cho, Kyle Robert, Xiaoxiao Chen, Lian Shu, Guangxian Tang, Jihong Wu, Xinghuai Sun, and Dong F. Chen
- Subjects
Male ,0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,retinal ischemia/reperfusion ,visual acuity ,T cell ,Immunology ,microglia ,Receptor, Macrophage Colony-Stimulating Factor ,Retina ,Rats, Sprague-Dawley ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Humans ,Immunology and Allergy ,Optic Neuropathy, Ischemic ,retinal ganglion cell ,Receptor ,positive scotopic threshold response ,Original Research ,Microglia ,business.industry ,neurodegeneration ,Retinal ,Recovery of Function ,medicine.disease ,colony stimulating factor-1 ,Antibodies, Neutralizing ,Rats ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Cellular Microenvironment ,chemistry ,Retinal ganglion cell ,business ,lcsh:RC581-607 ,Reperfusion injury ,030215 immunology - Abstract
Retinal ischemia/reperfusion injury (RI) is a common cause of irreversible visual impairment and blindness in elderly and critical unmet medical need. While no effective treatment is available for RI, microglial activation and local immune responses in the retina are thought to play important roles in the pathophysiology of neurodegeneration. While survival and activation of microglia depend critically on colony-stimulating factor receptor (CSF-1R) signaling, it remains unclear if targeting the retinal immune microenvironments by CSF-1RAb after RI is sufficient to rescue vision and present a potentially effective therapy. Here we used rodent models of RI and showed that retinal ischemia induced by acute elevation of intraocular pressure triggered an early activation of microglia and macrophages in the retina within 12 h. This was followed by lymphocyte infiltration and increased production of pro-inflammatory cytokines. Intravitreal injection of CSF-1R neutralizing antibody (CSF-1RAb) after RI significantly blocked microglial activation and the subsequent T cell recruitment. This also led to improved retinal ganglion cell survival and function measured by cell quantification and electroretinogram positive scotopic threshold responses, as well as increased visual acuity and contrast sensitivity as assessed by optomotor reflex-based assays, when compared to the isotype-treated control group. Moreover, the administration of CSF-1RAb efficiently attenuated inflammatory responses and activation of human microglia in culture, suggesting a therapeutic target with human relevance. These results, together with the existing clinical safety profiles, support that CSF-1RAb may present a promising therapeutic avenue for RI, a currently untreatable condition, by targeting microglia and the immune microenvironment in the retina to facilitate neural survival and visual function recovery.
- Published
- 2020
- Full Text
- View/download PDF
24. Pulmonary administration of a CSF-1R inhibitor alters the balance of tumor-associated macrophages and supports first-line chemotherapy in a lung cancer model
- Author
-
Hanming Zhang, Sandro R. P. da Rocha, Rashed M. Almuqbil, Sulaiman S. Alhudaithi, and Fatemah S. Sunbul
- Subjects
Lung Neoplasms ,Combination therapy ,medicine.medical_treatment ,Pharmaceutical Science ,Receptor, Macrophage Colony-Stimulating Factor ,02 engineering and technology ,030226 pharmacology & pharmacy ,Small Molecule Libraries ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antineoplastic Combined Chemotherapy Protocols ,Tumor-Associated Macrophages ,Tumor Microenvironment ,Medicine ,Animals ,Humans ,Lung cancer ,Lung ,Protein Kinase Inhibitors ,Cisplatin ,Chemotherapy ,Tumor microenvironment ,business.industry ,Macrophages ,Receptor Protein-Tyrosine Kinases ,021001 nanoscience & nanotechnology ,medicine.disease ,medicine.anatomical_structure ,Cancer research ,0210 nano-technology ,business ,Ex vivo ,medicine.drug - Abstract
Lung cancers remain the leading cause of cancer-related death in both men and women. Infiltrating immune cells in the tumor microenvironment (TME) play a critical role in the formation, progression, and the response of solid tumors to therapy, including in lung cancers. Clinical studies have established that tumor-associated macrophages (TAMs) and their phenotypical composition are critical immune infiltrates in the lung TME, with the abundance of the M2-like phenotype negatively correlating with patient survival. Colony-Stimulating Factor 1 (CSF-1) receptor (CSF-1R) is a type III protein tyrosine kinase receptor that plays an important role in the recruitment and differentiation of monocytes into tumor-promoting M2-like TAMs and their survival. In this work we evaluated the therapeutic potential of PLX 3397 (PLX), a small molecule CSF-1R inhibitor (CSF-1Ri), upon local lung administration in an immune-competent mouse model of lung cancer. The efficacy of local lung delivered PLX as single therapy was investigated first. As assessed by immunofluorescence of sections of lung tumor nodules, a statistically significant reduction in M2-like TAMs and an increase in M1-like TAMs was observed, thus leading to a shift in the (M1/M2) balance. Those changes in abundance of immune infiltrates correlated with a significant decrease in tumor burden when compared to control. When combined with systemically administered cisplatin (CIS) PLX treatment provided further benefits, leading to a significant decrease in tumor burden when compared to either PLX or CIS treatments alone, as measured by bioluminescence intensity (BLI) in vivo (thoracic area) and ex vivo (lung tissue). This combination therapy led to the most pronounced increase in M1/M2 ratio, followed by a significant decrease in M2-like TAMs with the CIS therapy. This work is clinically relevant as it demonstrates the potential of local lung administration of PLX to support standard of care chemotherapy for lung cancer management. This is important as the pulmonary route of administration is a plausible strategy for reducing the total dose of CSF-1Ris as the tissue of interest (lungs) can be locally targeted. Because the major off-target effect of CSF-1Ris is liver toxicity, reducing systemic concentration will support translation of those therapies, especially in combination with standard of care chemotherapy that has significant off-target toxicity and patient attrition itself. This work is scientifically relevant as we demonstrate for the first time that local administration of a CSF-1Ri to the lungs leads to a shift in the balance of TAMs in the TME of a model of lung tumor, adding to the sparse literature of CSF-1Ris related to lung cancers.
- Published
- 2020
25. Anti-c-fms Antibody Prevents Osteoclast Formation and Bone Resorption in Co-Culture of Osteoblasts and Osteoclast Precursors In Vitro and in Ovariectomized Mice
- Author
-
Fumitoshi Ohori, Adya Pramusita, Yasuhiko Nara, Aseel Marahleh, Ria Kinjo, Wei Ren Shen, Jiawei Qi, Itaru Mizoguchi, Hideki Kitaura, Takahiro Noguchi, and Saika Ogawa
- Subjects
0301 basic medicine ,Osteoporosis ,Osteoclasts ,lcsh:Chemistry ,Mice ,0302 clinical medicine ,Receptor ,lcsh:QH301-705.5 ,Spectroscopy ,reproductive and urinary physiology ,biology ,Chemistry ,Antibodies, Monoclonal ,Osteoblast ,hemic and immune systems ,Cell Differentiation ,General Medicine ,M-CSF ,Computer Science Applications ,medicine.anatomical_structure ,RANKL ,030220 oncology & carcinogenesis ,embryonic structures ,osteoclast ,Cancellous Bone ,Ovariectomized rat ,Female ,Injections, Intraperitoneal ,musculoskeletal diseases ,anti-c-fms antibody ,medicine.medical_specialty ,Ovariectomy ,Receptor, Macrophage Colony-Stimulating Factor ,OVX ,Catalysis ,Bone resorption ,Article ,Inorganic Chemistry ,03 medical and health sciences ,In vivo ,Osteoclast ,Internal medicine ,medicine ,Animals ,Physical and Theoretical Chemistry ,Bone Resorption ,Molecular Biology ,Osteoblasts ,Organic Chemistry ,RANK Ligand ,Osteoprotegerin ,X-Ray Microtomography ,medicine.disease ,biological factors ,Coculture Techniques ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,lcsh:Biology (General) ,lcsh:QD1-999 ,biology.protein - Abstract
Osteoporosis morphology is characterized by bone resorption and decreases in micro-architecture parameters. Anti-osteoporosis therapy targets osteoclasts because bone resorption is a unique function of osteoclasts. Anti-c-fms antibodies against the receptor for macrophage colony-stimulating factor (M-CSF) inhibit osteoclast formation and bone resorption in vitro and in vivo. However, the effect of anti-c-fms antibodies on bone resorption in ovariectomized (OVX) mice is unknown. In this study, we evaluated the effect of anti-c-fms antibodies on osteoclast formation and bone resorption in osteoblast&ndash, osteoclast precursor co-culture in vitro and in OVX mice. Osteoblast and osteoclast precursor co-cultures treated with anti-c-fms antibodies showed significantly inhibited osteoclast formation, while cultures without anti-c-fms antibody treatment showed osteoclast formation. However, anti-c-fms antibodies did not change the receptor activator of nuclear factor kappa-B ligand (RANKL) or osteoprotegrin (OPG) expression during osteoblast and osteoclast differentiation in vitro. These results indicate that anti-c-fms antibodies directly affected osteoclast formation from osteoclast precursors in co-culture. OVX mice were treated with intraperitoneal injections of anti-c-fms antibody. The trabecular bone structure of the femur was assessed by micro-computer tomography. The anti-c-fms antibody inhibited osteoclast formation and bone loss compared with PBS-treated OVX mice. These results indicate potential for the therapeutic application of anti-c-fms antibodies for postmenopausal osteoporosis.
- Published
- 2020
26. An Early Microglial Response Is Needed To Efficiently Control Herpes Simplex Virus Encephalitis
- Author
-
Guy Boivin, Olus Uyar, Jocelyne Piret, Nataly Laflamme, Serge Rivest, Marie-Christine Venable, Karima Zarrouk, and Julie Carbonneau
- Subjects
Central Nervous System ,Male ,Chemokine ,medicine.medical_treatment ,Immunology ,Central nervous system ,Receptor, Macrophage Colony-Stimulating Factor ,Herpesvirus 1, Human ,Biology ,medicine.disease_cause ,Microbiology ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Phagocytosis ,Interferon ,Virology ,medicine ,Animals ,Neuroinflammation ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Mice, Inbred BALB C ,Microglia ,CD68 ,Macrophage Colony-Stimulating Factor ,Brain ,Viral Load ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,Herpes simplex virus ,medicine.anatomical_structure ,Cytokine ,Receptors, Granulocyte-Macrophage Colony-Stimulating Factor ,Insect Science ,biology.protein ,Cytokines ,Pathogenesis and Immunity ,Encephalitis, Herpes Simplex ,Chemokines ,030217 neurology & neurosurgery ,medicine.drug - Abstract
The role of a signaling pathway through macrophage colony-stimulating factor (MCSF) and its receptor, macrophage colony-stimulating factor 1 receptor (CSF1R), during experimental herpes simplex virus 1 (HSV-1) encephalitis (HSE) was studied by two different approaches. First, we evaluated the effect of stimulation of the MCSF/CSF1R axis before infection. Exogenous MCSF (40 μg/kg of body weight intraperitoneally [i.p.]) was administered once daily to BALB/c mice on days 4 and 2 before intranasal infection with 2,500 PFU of HSV-1. MCSF treatment significantly increased mouse survival compared to saline (50% versus 10%; P = 0.0169). On day 6 postinfection (p.i.), brain viral titers were significantly decreased, whereas beta interferon (IFN-β) was significantly increased in mice treated with MCSF compared to mice treated with saline. The number of CD68(+) (a phagocytosis marker) microglial cells was significantly increased in MCSF-treated mice compared to the saline-treated group. Secondly, we conditionally depleted CSF1R on microglial cells of CSF1R-loxP-CX3CR1-cre/ERT2 mice (in a C57BL/6 background) through induction with tamoxifen. The mice were then infected intranasally with 600,000 PFU of HSV-1. The survival rate of mice depleted of CSF1R (knockout [KO] mice) was significantly lower than that of wild-type (WT) mice (0% versus 67%). Brain viral titers and cytokine/chemokine levels were significantly higher in KO than in WT animals on day 6 p.i. Furthermore, increased infiltration of monocytes into the brains of WT mice was seen on day 6 p.i., but not in KO mice. Our results suggest that microglial cells are essential to control HSE at early stages of the disease and that the MCSF/CSF1R axis could be a therapeutic target to regulate their response to infection. IMPORTANCE Microglia appear to be one of the principal regulators of neuroinflammation in the central nervous system (CNS). An increasing number of studies have demonstrated that the activation of microglia could result in either beneficial or detrimental effects in different CNS disorders. Hence, the role of microglia during herpes simplex virus encephalitis (HSE) has not been fully characterized. Using experimental mouse models, we showed that an early activation of the MCSF/CSF1R axis improved the outcome of the disease, possibly by inducing a proliferation of microglia. In contrast, depletion of microglia before HSV-1 infection worsened the prognosis of HSE. Thus, an early microglial response followed by sustained infiltration of monocytes and T cells into the brain seem to be key components for a better clinical outcome. These data suggest that microglia could be a potential target for immunomodulatory strategies combined with antiviral therapy to better control the outcome of this devastating disease.
- Published
- 2020
27. Hedgehog Inhibitors Suppress Osteoclastogenesis in In Vitro Cultures, and Deletion of
- Author
-
Yukihiro, Kohara, Ryuma, Haraguchi, Riko, Kitazawa, Yuuki, Imai, and Sohei, Kitazawa
- Subjects
musculoskeletal diseases ,Male ,Aging ,Cell Survival ,Pyridines ,GLI2 ,GLI1 ,Osteoclasts ,Receptor, Macrophage Colony-Stimulating Factor ,Hedgehog signaling ,Thiophenes ,Zinc Finger Protein Gli2 ,Zinc Finger Protein GLI1 ,Article ,GANT-58 ,Mice ,Osteogenesis ,GANT-61 ,Animals ,Hedgehog Proteins ,cyclopamine ,Cells, Cultured ,Mice, Knockout ,Smoothened ,Macrophages ,Veratrum Alkaloids ,Gene Expression Regulation, Developmental ,X-Ray Microtomography ,Smoothened Receptor ,Up-Regulation ,Mice, Inbred C57BL ,Pyrimidines ,Osteoporosis ,Signal Transduction - Abstract
The functional role of the Hedgehog (Hh)-signaling pathway has been widely investigated in bone physiology/development. Previous studies have, however, focused primarily on Hh functions in bone formation, while its roles in bone resorption have not been fully elucidated. Here, we found that cyclopamine (smoothened (Smo) inhibitor), GANT-58 (GLI1 inhibitor), or GANT-61 (GLI1/2 inhibitor) significantly inhibited RANKL-induced osteoclast differentiation of bone marrow-derived macrophages. Although the inhibitory effects were exerted by cyclopamine or GANT-61 treatment during 0–48 h (early stage of osteoclast differentiation) or 48–96 h (late stage of osteoclast differentiation) after RANKL stimulation, GANT-58 suppressed osteoclast formation only during the early stage. These results suggest that the Smo-GLI1/2 axis mediates the whole process of osteoclastogenesis and that GLI1 activation is requisite only during early cellular events of osteoclastogenesis. Additionally, macrophage/osteoclast-specific deletion of Smo in mice was found to attenuate the aging phenotype characterized by trabecular low bone mass, suggesting that blockage of the Hh-signaling pathway in the osteoclast lineage plays a protective role against age-related bone loss. Our findings reveal a specific role of the Hh-signaling pathway in bone resorption and highlight that its inhibitors show potential as therapeutic agents that block osteoclast formation in the treatment of senile osteoporosis.
- Published
- 2020
28. Novel Class of Colony-Stimulating Factor 1 Receptor Kinase Inhibitors Based on an
- Author
-
Zhicheng, Xie, Bing, Wu, Yingqiang, Liu, Wenming, Ren, Linjiang, Tong, Caigui, Xiang, Aihuan, Wei, Yuanzhuo, Gao, Limin, Zeng, Hua, Xie, Wei, Tang, and Youhong, Hu
- Subjects
Inflammation ,Lipopolysaccharides ,Molecular Structure ,Pyridines ,Anti-Inflammatory Agents ,Receptor, Macrophage Colony-Stimulating Factor ,Rats, Sprague-Dawley ,Small Molecule Libraries ,Mice ,Structure-Activity Relationship ,RAW 264.7 Cells ,Alkynes ,Animals ,Female ,Protein Kinase Inhibitors - Abstract
Colony-stimulating factor 1 receptor (CSF-1R) is involved in inflammatory disorders as well as in many types of cancer. Based on high-throughput screening and docking results, we performed a detailed structure-activity-relationship study, leading to the discovery of a new series of compounds with nanomolar IC
- Published
- 2020
29. Targeting myeloid-inflamed tumor with anti-CSF-1R antibody expands CD137+ effector T-cells in the murine model of pancreatic cancer
- Author
-
May Tun Saung, Alex B. Blair, Elizabeth M. Jaffee, Ding Ding, Dwayne L. Thomas, Takahiro Tsujikawa, Lei Zheng, Stephen Muth, and Lisa M. Coussens
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Myeloid ,Dendritic cells ,Pancreatic ductal adenocarcinoma ,Mice ,0302 clinical medicine ,CD137 ,PD-1 ,Immunology and Allergy ,Cytotoxic T cell ,Lymphoid aggregates ,biology ,Tumor associated macrophages ,CD28 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,GVAX ,3. Good health ,Cytotoxic T-cells ,Survival Rate ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Research Article ,CSF-1R ,Carcinoma, Pancreatic Ductal ,Combination therapy ,CD3 ,Immunology ,Receptor, Macrophage Colony-Stimulating Factor ,Adenocarcinoma ,lcsh:RC254-282 ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,03 medical and health sciences ,Pancreatic cancer ,medicine ,Animals ,Humans ,Pharmacology ,business.industry ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,biology.protein ,Interferon-γ ,business - Abstract
Background The pancreatic cancer vaccine, GVAX, induces novel lymphoid aggregates in the otherwise immune quiescent pancreatic ductal adenocarcinoma (PDAC). GVAX also upregulates the PD-1/PD-L1 pathway, and a pre-clinical model demonstrated the anti-tumor effects of combination GVAX and anti-PD-1 antibody therapy (GVAX/αPD-1). Resistance to GVAX was associated with an immune-suppressive myeloid cell infiltration, which may limit further therapeutic gains of GVAX/αPD-1 therapy. The expression of CSF-1R, a receptor important for myeloid cell migration, differentiation and survival, and the effect of its therapeutic blockade in the context of GVAX in PDAC has not been investigated. Methods Lymphoid aggregates appreciated in 24 surgically resected PDAC from patients who received one dose of neoadjuvant GVAX were analyzed with multiplex immunohistochemistry. Flow cytometry analysis of tumor infiltrating T-cells in a murine model of PDAC was performed to investigate the therapeutic effects and mechanism of anti-CSF-1R/anti-PD-1/GVAX combination immunotherapy. Results High CSF-1R expression in resected PDAC from patients who received neoadjuvant GVAX was associated with a higher myeloid to lymphoid cell ratio (p
- Published
- 2018
- Full Text
- View/download PDF
30. A reduced M1-like/M2-like ratio of macrophages in healthy adipose tissue expansion during SGLT2 inhibition
- Author
-
Kentaro Mori, Yoshihiro Ogawa, Naomi Ogasawara, Chikara Komiya, Kyoichiro Tsuchiya, Kumiko Shiba, and Yasutaka Miyachi
- Subjects
Male ,0301 basic medicine ,Adipose tissue ,lcsh:Medicine ,Ketone Bodies ,Mice ,0302 clinical medicine ,Glucosides ,lcsh:Science ,Interleukin-15 ,Mice, Knockout ,Multidisciplinary ,Chemistry ,Intracellular Signaling Peptides and Proteins ,Cell Polarity ,Interleukin ,Lipids ,Sphingomyelins ,Up-Regulation ,Phenotype ,Adipose Tissue ,Lipogenesis ,Phosphoenolpyruvate Carboxykinase (GTP) ,medicine.symptom ,Signal Transduction ,medicine.medical_specialty ,Stromal cell ,Receptors, CCR2 ,Adipose tissue macrophages ,Down-Regulation ,Receptor, Macrophage Colony-Stimulating Factor ,030209 endocrinology & metabolism ,Inflammation ,Thiophenes ,Anisoles ,Carbohydrate metabolism ,Ceramides ,Diet, High-Fat ,Article ,03 medical and health sciences ,Sodium-Glucose Transporter 2 ,3T3-L1 Cells ,Internal medicine ,medicine ,Animals ,Sodium-Glucose Transporter 2 Inhibitors ,Macrophages ,lcsh:R ,Lipid metabolism ,Mice, Inbred C57BL ,Pyrimidines ,030104 developmental biology ,Endocrinology ,lcsh:Q - Abstract
The adipose tissue includes various stromal cells, such as preadipocytes, endothelial cells, fibroblasts, and immune cells, which are involved in adipose tissue functions. We previously reported that, in obese mice, the sodium–glucose cotransporter 2 inhibitor ipragliflozin (Ipra) promoted the expansion of the epididymal adipose tissue (Epi) with increase of serum ketone body concentration. The Ipra-induced adipose tissue expansion did not deteriorate adipose inflammation, or systemic glucose/lipid metabolism, referred to as “healthy adipose tissue expansion.” Here we found that Ipra promoted healthy adipose tissue expansion with a reduced ratio of pro-inflammatory M1-like adipose tissue macrophages (ATMs) to anti-inflammatory M2-like ATMs. Ipra downregulated the gene expression of interleukin (IL)−15 (Il15) in stromal cells of Epi. IL-15 inhibited lipogenesis in 3T3-L1 cells associated with downregulation of the lipogenic gene. Ketone body β-hydroxybutyrate suppressed Il15 gene induction in M1-polarized cultured macrophages, and a ketogenic diet reproduced the adipose tissue expansion without deteriorating systemic glucose metabolism in mice. Our data indicate that the phenotypic switch of ATMs could mediate healthy adipose tissue expansion by treatment with Ipra, and it may offer new insights into the pathophysiological mechanisms of adipose tissue expansion.
- Published
- 2018
- Full Text
- View/download PDF
31. Bone marrow mesenchymal stem cells tune the differentiation of myeloid-derived suppressor cells in bleomycin-induced lung injury
- Author
-
Jun Wang, Lu Yu, Jie Huang, KaiHua Zhou, YinYin Wang, Yu Fang, Fei Qiao, Xiaosan Su, Yan-feng Yin, and Liu Yang
- Subjects
0301 basic medicine ,Neutrophils ,Integrin alpha4 ,Medicine (miscellaneous) ,Bone Marrow Cells ,Receptor, Macrophage Colony-Stimulating Factor ,Lung injury ,CD49d ,Bleomycin ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Monocytes ,lcsh:Biochemistry ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Bone marrow mesenchymal stem cells ,0302 clinical medicine ,Animals ,Humans ,Macrophage ,Cell Lineage ,Myeloid Cells ,lcsh:QD415-436 ,Cell Proliferation ,lcsh:R5-920 ,CD11b Antigen ,biology ,Research ,Cell Differentiation ,Mesenchymal Stem Cells ,Lung Injury ,Cell Biology ,Fibrosis ,In vitro ,030104 developmental biology ,chemistry ,Integrin alpha M ,Myeloid-derived suppressor cells ,030220 oncology & carcinogenesis ,Cancer research ,Myeloid-derived Suppressor Cell ,biology.protein ,Molecular Medicine ,Stem cell ,lcsh:Medicine (General) - Abstract
Background Bone marrow mesenchymal stem cells (BMSC) transfer has been attempted as a therapeutic strategy in experimental lung injury and fibrosis. Reduction of neutrophilic infiltration is one of the mechanisms involved in this effect. However, the mechanisms by which BMSC modulate neutrophil remains unknown. Methods and results Exposure of mice to bleomycin (BLM) resulted in significant accumulation of cells that express neutrophilic markers Gr-1HighCD11b+Ly-6GHighF4/80―CD115―CD49d―. These cells lacked immunosuppressive activity and could not be defined as myeloid-derived suppressor cells (MDSC). When BMSC were administrated to BLM-treated mice, they tuned the differentiation of Gr-1HighCD11b+ toward Gr-1LowCD11b+ cells. Gr-1LowCD11b+ cells exhibited unsegmented nuclei and expressed F4/80, Ly-6C, CD49d, and CD115 markers. These cells had potent immunosuppressive activity and thus could be defined as monocytic MDSC. As a result of such immunoregulation, BMSC mediated a decrease of pro-inflammatory products and amelioration of lung injury in BLM-treated mice. Further study using antibody array showed increased expression of macrophage colony-stimulating factor (M-CSF) in BMSC-treated mice. Accumulation of Gr-1LowCD11b+ cells in BMSC-treated mice was abrogated in M-CSF neutralizing mice. The beneficial effect of BMSC was independent of the ability of the cells to engraft in lung and in vitro coculture study of BMSC with Gr-1+CD11b+ cells showed that the induction of Gr-1LowCD11b+ cells by BMSC was independent of cell-cell contact. Conclusions These results document the generation of Gr-1HighCD11b+ cells in BLM-treated mice, and suggest that BMSC tune the differentiation of Gr-1HighCD11b+ toward Gr-1LowCD11b+ cells and therefore inhibit the progression of BLM-induced lung injury. Electronic supplementary material The online version of this article (10.1186/s13287-018-0983-1) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
32. Microglia: Same same, but different
- Author
-
Marco Prinz and Katrin Kierdorf
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,Immunology ,Central nervous system ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,News ,Motor Activity ,Insights ,Colony stimulating factor 1 receptor ,03 medical and health sciences ,Mice ,Purkinje Cells ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Receptor ,Social Behavior ,Microglia ,Behavior, Animal ,Macrophage Colony-Stimulating Factor ,Colony-stimulating factor ,030104 developmental biology ,medicine.anatomical_structure ,Signal transduction ,Neuroscience ,030217 neurology & neurosurgery ,Homeostasis ,Signal Transduction - Abstract
Kana et al. elegantly show that CSF-1, and not IL-34, orchestrates cerebellar microglial homeostasis in mice., Microglial identity in the central nervous system (CNS) is dependent on colony stimulating factor 1 receptor (CSF-1R) signaling and its ligands IL-34 and colony stimulating factor 1 (CSF-1). In this issue of JEM, Kana et al. (https://doi.org/10.1084/jem.20182037) make the important discovery that CSF-1, but not IL-34, orchestrates cerebellar microglial homeostasis in mice, and its deficiency resulted in severe cerebellar dysfunctions accompanied by defects in motor function and social behavior.
- Published
- 2019
33. Effect of CSF1R inhibitor on glial cells population and remyelination in the cuprizone model
- Author
-
Iraj Ragerdi Kashani, Shirin Barati, and Fatemeh Tahmasebi
- Subjects
Male ,Multiple Sclerosis ,Animal food ,Population ,Aminopyridines ,Receptor, Macrophage Colony-Stimulating Factor ,Microgliosis ,Luxol fast blue stain ,Colony stimulating factor 1 receptor ,Andrology ,Cuprizone ,Mice ,Cellular and Molecular Neuroscience ,Endocrinology ,medicine ,Animals ,Pyrroles ,Remyelination ,education ,Myelin Sheath ,education.field_of_study ,Endocrine and Autonomic Systems ,business.industry ,General Medicine ,Oligodendrocyte ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Neurology ,business ,Neuroglia ,Astrocyte - Abstract
Multiple sclerosis is a kind of autoimmune and demyelinating disease and its pathological symptoms include inflammation, myelin loss, astrocytosis, and microgliosis. The colony stimulating factor 1 receptor (CSF1R), as an essential factor for the microglial function, and PLX3397 is its specific inhibitor. In this study, we assessed the effect of different doses of PLX3397 for microglial ablation on glial cells population and remyelination process. Sixty male C57BL/6 mice (8 weeks old) were divided into 6 groups. The animals were fed with 0.2% cuprizone diet for 12 weeks for chronic demyelination model induction. For microglial ablation, PLX3397 (290 mg/kg) was added to the animal food for 3, 7, 14 and 21 days. Glial cells number was measured using immunohistochemistry. We evaluated the rate of remyelination using electron microscopy and Luxol Fast Blue staining. The expression levels of all genes were assessed by qRT-PCR method. Data were analysed using Graph pad prism and SPSS software.Data showed that the administration of different doses of PLX3397 significantly (p˂0.001) reduced microglial cells. PLX3397 administration significantly (p˂0.001) increased oligodendrocytes population with along rise of the remyelination compared to the cuprizone mice, which was confirmed by the results of LFB and TEM. Gene results showed that PLX3397 treatment reduced CSF1R expression. According to results, the administration of PLX3397 for 21 days enhanced remyelination by increasing oligodendrocytes in the chronic demyelination model. These positive effects could be related to the reduction of microglia.
- Published
- 2021
- Full Text
- View/download PDF
34. Combination of CD40 Agonism and CSF-1R Blockade Reconditions Tumor-Associated Macrophages and Drives Potent Antitumor Immunity
- Author
-
Michael Quigley, Douglas H. Yamada, Raluca Verona, Szeman Ruby Chan, Andressa Smith, Natasha M. Girgis, Iqbal S. Grewal, and Karla R. Wiehagen
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Immunology ,Priming (immunology) ,Antineoplastic Agents ,Receptor, Macrophage Colony-Stimulating Factor ,CD8-Positive T-Lymphocytes ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,Immune system ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Myeloid Cells ,CD40 Antigens ,Tumor microenvironment ,CD40 ,biology ,Macrophages ,FOXP3 ,Tumor antigen ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,Cytokines ,Female - Abstract
Efficacious antitumor immune responses must overcome multiple suppressive mechanisms in the tumor microenvironment to control cancer progression. In this study, we demonstrate that dual targeting of suppressive myeloid populations by inhibiting CSF-1/CSF-1R signaling and activation of antigen-presenting cells with agonist anti-CD40 treatment confers superior antitumor efficacy and increased survival compared with monotherapy treatment in preclinical tumor models. Concurrent CSF-1R blockade and CD40 agonism lead to profound changes in the composition of immune infiltrates, causing an overall decrease in immunosuppressive cells and a shift toward a more inflammatory milieu. Anti-CD40/anti–CSF-1R–treated tumors contain decreased tumor-associated macrophages and Foxp3+ regulatory T cells. This combination approach increases maturation and differentiation of proinflammatory macrophages and dendritic cells and also drives potent priming of effector T cells in draining lymph nodes. As a result, tumor-infiltrating effector T cells exhibit improved responses to tumor antigen rechallenge. These studies show that combining therapeutic approaches may simultaneously remove inhibitory immune populations and sustain endogenous antitumor immune responses to successfully impair cancer progression. Cancer Immunol Res; 5(12); 1109–21. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
35. Macrophage colony-stimulating factor (M-CSF) is an intermediate in the process of luteinizing hormone-induced decrease in natriuretic peptide receptor 2 (NPR2) and resumption of oocyte meiosis
- Author
-
Ying Feng, Chang Liu, Zhifen Zhang, Wenchao Sun, Xiaoyang Fei, Guangchao Zhuo, and Wen-Jing Zhou
- Subjects
0301 basic medicine ,Luteinizing hormone ,medicine.medical_specialty ,endocrine system ,Oocyte ,media_common.quotation_subject ,Gene Expression ,Receptor, Macrophage Colony-Stimulating Factor ,Natriuretic peptide receptor 2 ,lcsh:Gynecology and obstetrics ,Human chorionic gonadotropin ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Ovarian Follicle ,Internal medicine ,medicine ,Animals ,Meiosis resumption ,RNA, Messenger ,Ovarian follicle ,Equine chorionic gonadotropin ,Ovulation ,lcsh:RG1-991 ,media_common ,030219 obstetrics & reproductive medicine ,Germinal vesicle ,urogenital system ,Chemistry ,Macrophage colony-stimulating factor ,Research ,Ovary ,Obstetrics and Gynecology ,NPR2 ,Meiosis ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Oncology ,Oocytes ,Female ,Receptors, Atrial Natriuretic Factor ,Signal Transduction - Abstract
Background Luteinizing hormone (LH) regulation of the ligand, natriuretic peptide precursor type C, and its receptor, natriuretic peptide receptor 2 (NPR2), is critical for oocyte maturation; however, the mechanism is not fully understood. Macrophage colony-stimulating factor (M-CSF) has recently been shown to be involved in oocyte maturation and ovulation. In the present study we determined whether or not M-CSF plays a role in the intermediate signal that mediates LH regulation of NPR2 in resumption of oocyte meiosis. Methods Immature female C57BL/6 mice were injected i.p. with 5 IU of equine chorionic gonadotropin (eCG) to stimulate follicle development. After 44–48 h, the eCG-stimulated mice were injected i.p. with an ovulatory dose of 5 IU of human chorionic gonadotropin (hCG). The ovaries were excised at selected times. Pre-ovulatory follicles (POFs) and cumulus-oocyte complexes were cultured in different media. Immunohistochemical and quantitative real-time PCR analyses were used to assess the expression of M-CSF, M-CSF receptor (M-CSF-R), and NPR2. The presence of germinal vesicle breakdown (GVBD) was examined under a stereomicroscope to morphologically evaluate resumption of oocyte meiosis. Results NPR2 was mainly expressed in cumulus cells of pre-ovulatory follicles, while M-CSF and M-CSF-R were expressed in both mural granulosa and cumulus cells. The levels of M-CSF/M-CSF-R and NPR2 decreased within 4 h after treatment of hCG. M-CSF not only reduced the expression of NPR2 mRNA via its receptor (M-CSF-R), but also increased the proportion of GVBD in oocytes. Conclusion M-CSF serves as an intermediate signal, thus inducing a vital decrease in the NPR2 levels in cumulus cells, and regulates the process of LH-induced resumption of meiosis.
- Published
- 2017
- Full Text
- View/download PDF
36. Neuromedin B and its receptor silencing suppresses osteoclast generation by modulating precursor proliferation via M-CSF/c-Fms/D-type cyclins
- Author
-
Young-Ran Yoon, Seung Il Cho, Hyun-Ju Kim, Hae Won Lee, Sook Jin Seong, Chae-Eun Yeo, and Woo Youl Kang
- Subjects
musculoskeletal diseases ,0301 basic medicine ,MAPK/ERK pathway ,Neurokinin B ,Down-Regulation ,Osteoclasts ,Apoptosis ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,Small hairpin RNA ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteoclast ,Cyclin D ,medicine ,Animals ,Gene silencing ,Cell Lineage ,Gene Silencing ,Protein kinase B ,Cell Proliferation ,Gene knockdown ,Macrophage Colony-Stimulating Factor ,Stem Cells ,RANK Ligand ,Cell Differentiation ,Cell Biology ,Neuromedin B ,Receptors, Bombesin ,030104 developmental biology ,medicine.anatomical_structure ,RANKL ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Signal Transduction - Abstract
Neuromedin B (NMB), a mammalian bombesin-like peptide, regulates diverse physiological processes, such as energy metabolism, memory and fear behavior, and cellular growth, through its cognate receptor, NMBR. In this study, we report that NMB expression was upregulated during osteoclast development and that silencing NMB or NMBR attenuated osteoclast generation mediated by macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). We found that knockdown of NMB or NMBR using a small hairpin RNA suppressed M-CSF-induced proliferation of osteoclast precursor cells without altering osteoclast differentiation. Interestingly, NMB or NMBR knockdown reduced the expression of the M-CSF receptor, c-Fms, which is an important modulator of osteoclast development. Consequently, NMB or NMBR silencing inhibited M-CSF/c-Fms-mediated downstream signaling pathways like activation of ERK and Akt and induction of D-type cyclins, cyclin D1 and D2. Moreover, knockdown of NMB or NMBR accelerated apoptosis in osteoclast lineage cells by inducing caspase-3, caspase-9, and Bim expression. In summary, our study demonstrates that the NMB/NMBR axis plays a pivotal role in osteoclast generation by modulating the proliferation and survival of osteoclast lineage cells.
- Published
- 2017
- Full Text
- View/download PDF
37. Novel Microglia Depletion Systems: A Genetic Approach Utilizing Conditional Diphtheria Toxin Receptor Expression and a Pharmacological Model Based on the Blocking of Macrophage Colony-Stimulating Factor 1 Receptor
- Author
-
Maja, Kitic, Peter, See, Julia, Bruttger, Florent, Ginhoux, and Ari, Waisman
- Subjects
Mice ,CX3C Chemokine Receptor 1 ,Animals ,Gene Expression ,Diphtheria Toxin ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Microglia ,Antibodies, Blocking ,Promoter Regions, Genetic ,Heparin-binding EGF-like Growth Factor - Abstract
Microglia are the main population of macrophage residing in the central nervous system (CNS). Depletion experiments gave important insights into the physiology and function of microglia in healthy and diseased CNS. Ablation of microglia can be achieved by application of pharmacological or genetic tools. Here, we describe two approaches to ablate microglia: an efficient genetic model that utilizes DTR
- Published
- 2019
38. CSF-1 receptor inhibition as a highly effective tool for depletion of microglia in mixed glial cultures
- Author
-
Asparouh I. Iliev and Sabrina Hupp
- Subjects
0301 basic medicine ,Lipopolysaccharide ,Receptor, Macrophage Colony-Stimulating Factor ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,medicine ,Receptor Inhibition ,Macrophage ,Animals ,610 Medicine & health ,Cells, Cultured ,Microglia ,General Neuroscience ,Cell preparation ,In vitro ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Decreased Sensitivity ,Astrocytes ,570 Life sciences ,biology ,Neuroglia ,030217 neurology & neurosurgery ,Homogenization (biology) - Abstract
Background A breakthrough in the microglia and macrophages field was the identification of the macrophage colony stimulating factor-1 (CSF-1) as a pro-survival factor. Its pharmacological inhibition in animals depletes rapidly all microglia and macrophages. Microglial depletion in mixed glial cultures has always represented a challenge and none of the existing approaches delivers satisfactory results. New Method We applied a CSF-1R inhibitor (PLX5622) in primary mouse glial cultures, analyzing microglial dose-responses, starting at different time-points and incubating for various periods of time. Results We used two treatment modalities with 10 μM PLX5622 to deplete microglia: i) immediately after brain homogenization and ii) at day in vitro 12. The application of the inhibitor immediately after cell preparation depleted microglia to 8% at 1 week, to 2% at 4 weeks and to 0.5% at 6 weeks (half-time 3.5 days). When mixed glial cultures were treated starting at day in vitro 12, microglia depletion was slower (half-time 6 days) and not complete, indicating a decreased sensitivity to CSF-1. The remaining astrocytes preserved their proliferation ability, their migration in a scratch wound assay, and their pro-inflammatory (IL-6) response towards lipopolysaccharide. Comparison to Existing Methods The proposed approach for microglial depletion in mixed glial cultures is more effective than other existing methods and is non-toxic to non-microglial cells. Conclusions CSF-1R inhibitors are effective tools for depleting microglia in mixed glial cultures. Longer maturation of the cultures leads to a diminished sensitivity of microglia towards CSF-1. Thus, the treatment should start as early as possible after glial culture preparation.
- Published
- 2019
39. Heat-killed Mycobacterium tuberculosis prime-boost vaccination induces myeloid-derived suppressor cells with spleen dendritic cell-killing capability
- Author
-
Eliana, Ribechini, Ina, Eckert, Andreas, Beilhack, Nelita, Du Plessis, Gerhard, Walzl, Ulrike, Schleicher, Uwe, Ritter, and Manfred B, Lutz
- Subjects
Mice, Knockout ,CD11b Antigen ,Myeloid-Derived Suppressor Cells ,T-Lymphocytes ,Vaccination ,Immunization, Secondary ,Nitric Oxide Synthase Type II ,Receptor, Macrophage Colony-Stimulating Factor ,Dendritic Cells ,Mycobacterium tuberculosis ,Nitric Oxide ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Animals ,Antigens, Ly ,Cytokines ,Tuberculosis ,Myeloid Cells ,Tuberculosis Vaccines ,Spleen ,Cell Proliferation ,Research Article - Abstract
Tuberculosis patients and mice infected with live Mycobacterium tuberculosis accumulate high numbers of myeloid-derived suppressor cells (MDSCs). Here, we hypothesized that dead M. tuberculosis vaccines also may induce MDSCs that could impair the efficacy of vaccination. We found that repeated injections of M. tuberculosis vaccines (heat-killed M. tuberculosis in incomplete Freund’s adjuvant, such as Montanide) but not single or control vaccines without M. tuberculosis strongly expanded CD11b(+) myeloid cells in the spleen, leading to T cell suppression of proliferation and killing ex vivo. Dead M. tuberculosis vaccination induced the generation of CD11b(+)Ly6C(hi)CD115(+) iNOS/Nos2(+) monocytic MDSCs (M-MDSCs) upon application of inflammatory or microbial activation signals. In vivo these M-MDSCs were positioned strategically in the splenic bridging channels and then positioned in the white pulp areas. Notably, within 6–24 hours, in a Nos2-dependent fashion, they produced NO to rapidly kill conventional and plasmacytoid DCs while, surprisingly, sparing T cells in vivo. Thus, we demonstrate that M. tuberculosis vaccine induced M-MDSCs do not directly suppress effector T cells in vivo but, instead, indirectly by killing DCs. Collectively, we demonstrate that M. tuberculosis booster vaccines induce M-MDSCs in the spleen that can be activated to kill DCs. Our data suggest that formation of MDSCs by M. tuberculosis vaccines should be investigated also in clinical trials.
- Published
- 2019
40. Depletion of tumor-associated macrophages enhances the anti-tumor effect of docetaxel in a murine epithelial ovarian cancer
- Author
-
Xiaofen Lu and Tengteng Meng
- Subjects
0301 basic medicine ,Immunology ,Antineoplastic Agents ,Apoptosis ,Receptor, Macrophage Colony-Stimulating Factor ,Docetaxel ,CD8-Positive T-Lymphocytes ,Carcinoma, Ovarian Epithelial ,Taxoid ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Cell Movement ,medicine ,Tumor Microenvironment ,Immunology and Allergy ,Animals ,Humans ,Epithelial ovarian cancer ,Benzothiazoles ,Neoplasm Metastasis ,Receptor ,Picolinic Acids ,Cell Proliferation ,Chemistry ,Macrophages ,Hematology ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Drug Resistance, Neoplasm ,Cancer research ,Drug Therapy, Combination ,Female ,Ovarian cancer ,Infiltration (medical) ,CD8 ,030215 immunology ,medicine.drug - Abstract
Docetaxel (DTXL), a new member of the taxoid family, has been used for cancer treatment. However, increasing cases of DTXL resistance have been reported. Tumor-associated macrophages (TAMs) have been implicated in tumor invasion and chemo-resistance. Eliminating TAMs by inhibiting colony stimulating factor-1 receptor (CSF-1R) has emerged as a promising strategy for cancer treatment. BLZ945 is a CSF-1R inhibitor and has anti-tumor function. In present study, anti-tumor effects of combination treatment of BLZ945 and DTXL were investigated. We established a mouse ovarian cancer model and investigated the effect of BLZ945, DTXL single treatment or combination treatment on TAMs infiltration, tumor growth, CD8+ T cell infiltration and cancer metastasis. DTXL treatment increased the infiltration while BLZ945 induced cell apoptosis in macrophages. DTXL/BLZ945 combination treatment significantly inhibited tumor growth, reduced the abundance of TAMs, increased CD8+ T cell infiltration and prevented lung metastasis. Depletion of Tumor-Associated Macrophages (TAMs) by BLZ945 enhanced the anti-tumor effect of DTXL in a murine epithelial ovarian cancer.
- Published
- 2019
41. Melanoma-conditioned medium promotes cytotoxic immune responses by murine bone marrow-derived monocytes despite their expression of 'M2' markers
- Author
-
Patrick F. Forde, Liam Friel Tremble, and Anne C. Moore
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,Skin Neoplasms ,Immunology ,Melanoma, Experimental ,Bone Marrow Cells ,Receptor, Macrophage Colony-Stimulating Factor ,Receptors, Cell Surface ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Th2 Cells ,Immunity ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Macrophage ,Animals ,Lectins, C-Type ,Melanoma ,Chemistry ,Macrophages ,Cell Differentiation ,Neoplasms, Experimental ,Coculture Techniques ,Granzyme B ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Mannose-Binding Lectins ,Oncology ,Culture Media, Conditioned ,Cancer research ,Cytokines ,Cytokine secretion ,Bone marrow ,CD8 ,Mannose Receptor ,030215 immunology - Abstract
Macrophages have been shown to infiltrate a wide range of malignancies and are often considered to promote tumour survival, growth and spread. However, the source and behaviour of discrete tumour-associated macrophage populations are still poorly understood. Here we show a novel method for the rational development of bone marrow-derived monocytes appropriate for the study of processes which involve the contribution of circulating inflammatory monocytes. We have shown that in response to tumour-conditioned medium, these cells upregulate CD206 and CD115, markers traditionally associated with M2-type macrophages. Treated cells show reduced capacity for cytokine secretion but significantly impact CD4+ and CD8+ T-cell proliferation and polarization. Coculture with conditioned bone marrow-derived monocytes significantly reduced CD4+ T-cell proliferation but increased CD8+ T-cell proliferation and granzyme B expression with significant induction of IFNγ secretion by both CD4+ and CD8+ T cells, indicating that these cells may have a role in promoting anti-cancer immunity.
- Published
- 2019
42. The CSF-1-receptor inhibitor, JNJ-40346527 (PRV-6527), reduced inflammatory macrophage recruitment to the intestinal mucosa and suppressed murine T cell mediated colitis
- Author
-
Jennifer E. Towne, Beverley A. Moore, Stanley M. Belkowski, Paul J. Dunford, S Plevy, Francisco Leon, Yanqing Chen, Xiang Yao, Matthew J. Loza, Hao Liu, Holly Raymond-Parks, and Carl L. Manthey
- Subjects
0301 basic medicine ,Pyridines ,T-Lymphocytes ,Gene Expression ,Crohn's Disease ,Inflammatory bowel disease ,Monocytes ,Mice ,White Blood Cells ,0302 clinical medicine ,Intestinal mucosa ,Animal Cells ,Medicine and Health Sciences ,Intestinal Mucosa ,Crohn's disease ,Multidisciplinary ,T Cells ,Imidazoles ,Animal Models ,Colitis ,medicine.anatomical_structure ,Experimental Organism Systems ,030220 oncology & carcinogenesis ,Medicine ,Tumor necrosis factor alpha ,Cellular Types ,Anatomy ,Signal Transduction ,Research Article ,Colon ,T cell ,Immune Cells ,Science ,Immunology ,Receptor, Macrophage Colony-Stimulating Factor ,Mouse Models ,Gastroenterology and Hepatology ,Research and Analysis Methods ,Autoimmune Diseases ,03 medical and health sciences ,Model Organisms ,medicine ,Genetics ,Animals ,Humans ,Protein Kinase Inhibitors ,Inflammation ,Blood Cells ,Dose-Response Relationship, Drug ,business.industry ,Microarray analysis techniques ,Gene Expression Profiling ,Macrophages ,Inflammatory Bowel Disease ,Biology and Life Sciences ,Cell Biology ,medicine.disease ,Gene expression profiling ,Gastrointestinal Tract ,030104 developmental biology ,Cancer research ,Animal Studies ,Clinical Immunology ,Clinical Medicine ,business ,Digestive System - Abstract
Background & aims Originally believed to be primarily a disorder of T-cell signaling, evidence shows that macrophage-lineage cells also contribute to the pathogenesis of Crohn's disease (CD). Colony stimulating factor-1 (CSF-1) is a key regulator of the macrophage lineage, but its role in CD has not been well established. We examined transcriptional data from CD mucosa for evidence of CSF-1 pathway activation and tested JNJ-40346527 (PRV-6527), a small molecule inhibitor of CSF-1 receptor kinase (CSF-1R), for its ability to inhibit disease indices in murine colitis. Methods A CSF-1 pathway gene set was created from microarray data of human whole blood cultured ex vivo with CSF-1 and compared to a TNFα-induced gene set generated from epithelial-lineage cells. Gene set variation analysis was performed using existing Crohn's mucosa microarray data comparing patients who either responded or failed to respond to anti-TNFα therapy. Commencing day 14 or day 21, mice with T-cell transfer colitis were treated with vehicle or JNJ-40346527 until study termination (day 42). Endpoints included colon weight/length ratios and histopathology scores, and macrophage and T cells were assessed by immunohistochemistry. Mucosal gene expression was investigated using RNAseq. Results Both the CSF-1 and the TNFα gene sets were enriched in the colonic mucosal transcriptomes of Crohn's disease and in mouse colitis, and expression of both gene sets was highest in patients who did not respond to anti-TNFα therapy. In these patients neither set was reduced by therapy. In the mouse model, JNJ-40346527 inhibited the increase in colon weight/length ratio by ∼50%, reduced histological disease scores by ∼60%, and reduced F4/80+ mononuclear cell and CD3+ lymphocyte numbers. RNAseq analysis confirmed the CSF-1 gene set was sharply reduced in treated mice, as were gene sets enriched in "M1" inflammatory and "M0" resident macrophages and in activated T cells. Conclusions CSF-1 biology is activated in Crohn's disease and in murine T cell transfer colitis. Inhibition of CSF-1R by JNJ-40346527 was associated with attenuated clinical disease scores and reduced inflammatory gene expression in mice. These data provide rationale for testing JNJ-40346527 (PRV-6527) in human inflammatory bowel disease.
- Published
- 2019
43. Establishment of an orthodontic retention mouse model and the effect of anti-c-Fms antibody on orthodontic relapse
- Author
-
Aseel Marahleh, Hideki Kitaura, Akiko Kishikawa, Yasuhiko Nara, Fumitoshi Ohori, Wei Ren Shen, Itaru Mizoguchi, Saika Ogawa, Jiawei Qi, and Takahiro Noguchi
- Subjects
0301 basic medicine ,Molar ,Male ,Teeth ,Dentistry ,Osteoclasts ,Mandibular first molar ,Mandibular second molar ,Mice ,0302 clinical medicine ,Osteogenesis ,Animal Cells ,Medicine and Health Sciences ,Medicine ,Musculoskeletal System ,Cells, Cultured ,Connective Tissue Cells ,Multidisciplinary ,biology ,Antibodies, Monoclonal ,Animal Models ,medicine.anatomical_structure ,Treatment Outcome ,Experimental Organism Systems ,Connective Tissue ,Antibody ,Tooth Mobility ,Anatomy ,Cellular Types ,Protein Binding ,Research Article ,Science ,Oral Medicine ,Receptor, Macrophage Colony-Stimulating Factor ,Orthodontics ,Mouse Models ,Molars ,Research and Analysis Methods ,03 medical and health sciences ,Model Organisms ,Osteoclast ,Animals ,Retention period ,Bone ,Immunohistochemistry Techniques ,Dental alveolus ,Skeleton ,business.industry ,Macrophage Colony-Stimulating Factor ,Skull ,Biology and Life Sciences ,030206 dentistry ,Cell Biology ,Histochemistry and Cytochemistry Techniques ,Disease Models, Animal ,030104 developmental biology ,Biological Tissue ,Jaw ,biology.protein ,Animal Studies ,Immunologic Techniques ,Alveolar Bone ,business ,Antibody therapy ,Digestive System ,Head - Abstract
Orthodontic relapse after orthodontic treatment is a major clinical issue in the dental field. However, the biological mechanism of orthodontic relapse is still unclear. This study aimed to establish a mouse model of orthodontic retention to examine how retention affects the rate and the amount of orthodontic relapse. We also sought to examine the role of osteoclastogenesis in relapse using an antibody to block the activity of M-CSF, an essential factor of osteoclast formation. Mice were treated with a nickel-titanium closed-coil spring that was fixed between the upper incisors and the upper-left first molar to move the first molar in a mesial direction over 12 days. Mice were randomly divided into three groups: group 1, no retention (G1); group 2, retention for 2 weeks (G2); and group 3, retention for 4 weeks (G3). In G2 and G3, a light-cured resin was placed in the space between the first and second molars as a model of retention. Orthodontic relapse was assessed by measuring changes in the dimensions of the gap created between the first and second molars. To assess the activity and role of osteoclasts, mice in G3 were injected with anti-c-Fms antibody or PBS, and assessed for changes in relapse distance and rate. Overall, we found that a longer retention period was associated with a slower rate of relapse and a shorter overall amount of relapse. In addition, inhibiting osteoclast formation using the anti-c-Fms antibody also reduced orthodontic relapse. These results suggest that M-CSF and/or its receptor could be potential therapeutic targets in the prevention and treatment of orthodontic relapse.
- Published
- 2019
44. Selective deletion of the receptor for CSF1, c-fms, in osteoclasts results in a high bone mass phenotype, smaller osteoclasts in vivo and an impaired response to an anabolic PTH regimen
- Author
-
Ben-hua Sun, Erin Nevius, João Pereira, Jared Kaplan, Meiling Zhu, and Karl L. Insogna
- Subjects
Male ,Anabolism ,Physiology ,medicine.medical_treatment ,Osteoclasts ,Immunostaining ,Mice ,Bone Density ,Osteogenesis ,Animal Cells ,Medicine and Health Sciences ,Cathepsin K ,Femur ,Receptor ,Musculoskeletal System ,Connective Tissue Cells ,Mice, Knockout ,Staining ,Multidisciplinary ,Chemistry ,Cell Staining ,Cell Differentiation ,medicine.anatomical_structure ,Cytokine ,Parathyroid Hormone ,Connective Tissue ,Cancellous Bone ,Knockout mouse ,Medicine ,Female ,Bone Remodeling ,Anatomy ,Cellular Types ,Research Article ,musculoskeletal diseases ,Macrophage colony-stimulating factor ,medicine.medical_specialty ,Science ,Receptor, Macrophage Colony-Stimulating Factor ,Research and Analysis Methods ,Osteocytes ,In vivo ,Osteoclast ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Bone Resorption ,Bone ,Skeleton ,Osteoblasts ,Biology and Life Sciences ,X-Ray Microtomography ,Cell Biology ,Peptide Fragments ,Mice, Inbred C57BL ,Biological Tissue ,Endocrinology ,Specimen Preparation and Treatment ,Physiological Processes - Abstract
The receptor for Colony Stimulating Factor 1 (CSF1), c-fms, is highly expressed on mature osteoclasts suggesting a role for this cytokine in regulating the function of these cells. Consistent with this idea, in vitro studies have documented a variety of effects of CSF1 in mature osteoclasts. To better define the role of CSF1 in these cells, we conditionally deleted c-fms in osteoclasts (c-fms-OC-/-) by crossing c-fmsflox/flox mice with mice expressing Cre under the control of the cathepsin K promoter. The c-fms-OC-/- mice were of normal weight and had normal tooth eruption. However, when quantified by DXA, bone mass was significantly higher in the spine and femur of female knock out mice and in the femurs of male knock out mice. MicroCT analyses of femurs showed that female c-fms-OC-/- mice had significantly increased trabecular bone mass with a similar trend in males and both sexes demonstrated significantly increased trabecular number and reduced trabecular spacing. Histomorphometric analysis of the femoral trabecular bone compartment demonstrated a trend towards increased numbers of osteoclasts, +26% in Noc/BPm and +22% in OcS/BS in the k/o animals but this change was not significant. However, when the cellular volume of osteoclasts was quantified, the c-fms-OC-/- cells were found to be significantly smaller than controls. Mature osteoclasts show a marked spreading response when exposed to CSF1 in a non-gradient fashion. However, osteoclasts freshly isolated from c-fms-OC-/- mice had a near complete abrogation of this response. C-fms-OC-/- mice treated with (1–34)hPTH 80 ng/kg/d in single daily subcutaneous doses for 29 days showed an attenuated anabolic response in trabecular bone compared to wild-type animals. Taken together, these data indicate an important non-redundant role for c-fms in regulating mature osteoclast function in vivo.
- Published
- 2021
- Full Text
- View/download PDF
45. Targeting Suppressive Myeloid Cells Potentiates Checkpoint Inhibitors to Control Spontaneous Neuroblastoma
- Author
-
John Inge Johnsen, Katarina Le Blanc, Per Kogner, Rolf Kiessling, Nina Eissler, and Yumeng Mao
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,CD34 ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Kaplan-Meier Estimate ,B7-H1 Antigen ,Monocytes ,Immunomodulation ,Mice ,Neuroblastoma ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Benzothiazoles ,Molecular Targeted Therapy ,Picolinic Acids ,Myelopoiesis ,business.industry ,Myeloid-Derived Suppressor Cells ,Immunotherapy ,Hematopoietic Stem Cells ,Prognosis ,medicine.disease ,Disease Models, Animal ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Immunology ,Bone marrow ,Stem cell ,business - Abstract
Purpose: Neuroblastoma is the most common extracranial solid cancer type in childhood, and high-risk patients have poor prognosis despite aggressive multimodal treatment. Neuroblastoma-driven inflammation contributes to the induction of suppressive myeloid cells that hamper efficient antitumor immune responses. Therefore, we sought to enhance antitumor immunity by removing immunosuppression mediated by myeloid cells. Experimental Design: The prognostic values of myeloid cells are demonstrated by analyzing genomic datasets of neuroblastoma patients. The impact of tumor-derived factors on myelopoiesis and local induction of suppressive myeloid cells is dissected by in vitro culture models using freshly isolated human CD34+ hematopoietic stem cells, primary human monocytes, and murine bone marrow cells. To test the therapeutic efficacy of BLZ945 as a monotherapy or in combination with checkpoint inhibitors, we used a transgenic murine model (TH-MYCN) that develops aggressive spontaneous neuroblastoma. Results: We report that infiltrating CSF-1R+ myeloid cells predict poor clinical outcome in patients with neuroblastoma. In vitro, neuroblastoma-derived factors interfere with early development of myeloid cells and enable suppressive functions on human monocytes through M-CSF/CSF-1R interaction. In a transgenic mouse model (TH-MYCN) resembling high-risk human neuroblastoma, antagonizing CSF-1R with a selective inhibitor (BLZ945) modulates the induction of human and murine suppressive myeloid cells and efficiently limit tumor progression. While checkpoint inhibitors are insufficient in controlling tumor growth, combining BLZ945 with PD-1/PD-L1 blocking antibodies results in superior tumor control. Conclusions: Our results demonstrate the essential role of CSF-1R signaling during the induction of suppressive myeloid cells and emphasize its clinical potential as an immunotherapy for human cancers. Clin Cancer Res; 22(15); 3849–59. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
46. Resident corneal c-fms + macrophages and dendritic cells mediate early cellular infiltration in adenovirus keratitis
- Author
-
Emma C. Materne, Mirja Ramke, Jaya Rajaiya, Xiaohong Zhou, and James Chodosh
- Subjects
0301 basic medicine ,Corneal Infection ,Adenoviridae Infections ,Chemokine CXCL1 ,Corneal Stroma ,Eye Infections, Viral ,Mice, Transgenic ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,Article ,Keratitis ,Cornea ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Macrophage ,Macrophages ,Dendritic Cells ,Dendritic cell ,medicine.disease ,Sensory Systems ,Mice, Inbred C57BL ,Cellular infiltration ,Disease Models, Animal ,Ophthalmology ,030104 developmental biology ,medicine.anatomical_structure ,Apoptosis ,Immunology ,030221 ophthalmology & optometry - Abstract
The cornea contains a heterogeneous population of antigen-presenting cells with the capacity to contribute to immune responses. Adenovirus keratitis is a severe corneal infection with acute and chronic phases. The role of resident corneal antigen-presenting cells in adenovirus keratitis has not been studied. We utilized transgenic MaFIA mice in which c-fms expressing macrophages and dendritic cells can be induced to undergo apoptosis, in a mouse model of adenovirus keratitis. Clinical keratitis and recruitment of myeloperoxidase and CD45(+) cells were diminished in c-fms depleted, adenovirus infected mice, as compared to controls, consistent with a role for myeloid-lineage cells in adenovirus keratitis.
- Published
- 2016
- Full Text
- View/download PDF
47. Targeting myeloid-derived suppressor cells with colony stimulating factor-1 receptor blockade can reverse immune resistance to immunotherapy in indoleamine 2,3-dioxygenase-expressing tumors
- Author
-
Taha Merghoub, Dmitriy Zamarin, Alexander M. Lesokhin, Jedd D. Wolchok, and Rikke B. Holmgaard
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Programmed Cell Death 1 Receptor ,Population ,Melanoma, Experimental ,MDSCs ,Receptor, Macrophage Colony-Stimulating Factor ,General Biochemistry, Genetics and Molecular Biology ,Receptor tyrosine kinase ,IDO ,Colony stimulating factor 1 receptor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,PD-1 ,Animals ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Medicine ,CTLA-4 Antigen ,Molecular Targeted Therapy ,Enzyme Inhibitors ,education ,Indoleamine 2,3-dioxygenase ,education.field_of_study ,biology ,business.industry ,Myeloid-Derived Suppressor Cells ,Drug Synergism ,General Medicine ,Immunotherapy ,Xenograft Model Antitumor Assays ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Drug Resistance, Neoplasm ,CTLA-4 ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Immunology ,biology.protein ,Myeloid-derived Suppressor Cell ,business ,Research Paper ,CSF-1R - Abstract
Tumor indoleamine 2,3-dioxygenase (IDO) promotes immunosuppression by direct action on effector T cells and Tregs and through recruitment, expansion and activation of myeloid-derived suppressor cells (MDSCs). Targeting of MDSCs is clinically being explored as a therapeutic strategy, though optimal targeting strategies and biomarkers predictive of response are presently unknown. Maturation and tumor recruitment of MDSCs are dependent on signaling through the receptor tyrosine kinase CSF-1R on myeloid cells. Here, we show that MDSCs are the critical cell population in IDO-expressing B16 tumors in mediating accelerated tumor outgrowth and resistance to immunotherapy. Using a clinically relevant drug, we show that inhibition of CSF-1R signaling can functionally block tumor-infiltrating MDSCs and enhance anti-tumor T cell responses. Furthermore, inhibition of CSF-1R sensitizes IDO-expressing tumors to immunotherapy with T cell checkpoint blockade, and combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. These findings provide evidence for a critical and functional role for MDSCs on the in vivo outcome of IDO-expressing tumors., Highlights • Tumor-infiltrating MDSCs promote accelerated outgrowth of IDO-expressing tumors. • MDSCs infiltrating IDO-expressing tumors mediate resistance to immunotherapy. • Combination of CSF-1R blockade with IDO inhibitors potently elicits tumor regression. • CSF-1R blockade sensitizes tumors to the effects of immune checkpoint blockade. Our data demonstrate that therapy with CSF-1R-blocking agents offers therapeutic benefit as a single agent and potentiates the effect of immunotherapeutic agents in IDO-expressing tumors infiltrated with CSF-1R-expressing MDSCs. These findings provide important insights into basic mechanisms underlying IDO mediated immune suppression and resistance to immunotherapies. In addition, it provides a strong rationale for therapeutic combinations with CSF-1R inhibitors in tumors expressing elevated IDO and highly infiltrated with MDSCs as predictive biomarkers.
- Published
- 2016
- Full Text
- View/download PDF
48. Perturbed MafB/GATA1 axis after burn trauma bares the potential mechanism for immune suppression and anemia of critical illness
- Author
-
Andrea Szilagyi, Richard L. Gamelli, Li K. He, Ravi Shankar, Joseph A. Posluszny, Kuzhali Muthumalaiappan, and Nicholas B. Johnson
- Subjects
Male ,0301 basic medicine ,Myeloid ,Transcription, Genetic ,Critical Illness ,Recombinant Fusion Proteins ,MafB Transcription Factor ,Immunology ,Receptor, Macrophage Colony-Stimulating Factor ,Plasmacytoid dendritic cell ,Biology ,Cell Development, Differentiation, & Trafficking ,Monocytes ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Immunology and Allergy ,Cell Lineage ,GATA1 Transcription Factor ,RNA, Small Interfering ,Progenitor cell ,Cells, Cultured ,Myeloid Progenitor Cells ,Myelopoiesis ,Macrophages ,Monocyte ,Anemia ,030208 emergency & critical care medicine ,Dendritic Cells ,Cell Biology ,Dendritic cell ,030104 developmental biology ,medicine.anatomical_structure ,MAFB ,RNA Interference ,Bone marrow ,Burns - Abstract
Patients who survive initial burn injury are susceptible to nosocomial infections. Anemia of critical illness is a compounding factor in burn patients that necessitates repeated transfusions, which further increase their susceptibility to infections and sepsis. Robust host response is dependent on an adequate number and function of monocytes/macrophages and dendritic cells. In addition to impaired RBC production, burn patients are prone to depletion of dendritic cells and an increase in deactivated monocytes. In steady-state hematopoiesis, RBCs, macrophages, and dendritic cells are all generated from a common myeloid progenitor within the bone marrow. We hypothesized in a mouse model of burn injury that an increase in myeloid-specific transcription factor V-maf musculoaponeurotic fibrosarcoma oncogene homolog B at the common myeloid progenitor stage steers their lineage potential away from the megakaryocyte erythrocyte progenitor production and drives the terminal fate of common myeloid progenitors to form macrophages vs. dendritic cells, with the consequences being anemia, monocytosis, and dendritic cell deficits. Results indicate that, even though burn injury stimulated bone marrow hematopoiesis by increasing multipotential stem cell production (LinnegSca1poscKitpos), the bone marrow commitment is shifted away from the megakaryocyte erythrocyte progenitor and toward granulocyte monocyte progenitors with corresponding alterations in peripheral blood components, such as hemoglobin, hematocrit, RBCs, monocytes, and granulocytes. Furthermore, burn-induced V-maf musculoaponeurotic fibrosarcoma oncogene homolog B in common myeloid progenitors acts as a transcriptional activator of M-CSFR and a repressor of transferrin receptors, promoting macrophages and inhibiting erythroid differentiations while dictating a plasmacytoid dendritic cell phenotype. Results from small interfering RNA and gain-of-function (gfp-globin transcription factor 1 retrovirus) studies indicate that targeted interventions to restore V-maf musculoaponeurotic fibrosarcoma oncogene homolog B/globin transcription factor 1 balance can mitigate both immune imbalance and anemia of critical illness.
- Published
- 2016
- Full Text
- View/download PDF
49. Src family kinase expression and subcellular localization in macrophages: implications for their role in CSF-1-induced macrophage migration
- Author
-
Amy R. Dwyer, Eloise L Greenland, Andrew Sunderland, Kellie A. Mouchemore, Natalia G. Sampaio, James H. Steer, David A. Joyce, and Fiona J. Pixley
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,Immunology ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,SH3 domain ,Mice ,03 medical and health sciences ,Cell Movement ,LYN ,Animals ,Humans ,Immunology and Allergy ,Src family kinase ,Phosphorylation ,Cells, Cultured ,Tyrosine-protein kinase CSK ,Kinase ,Macrophage Colony-Stimulating Factor ,Macrophages ,Cell Differentiation ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,src-Family Kinases ,030104 developmental biology ,Signal transduction ,Signal Transduction ,Subcellular Fractions ,Proto-oncogene tyrosine-protein kinase Src - Abstract
A major role of colony-stimulating factor-1 is to stimulate the differentiation of mononuclear phagocytic lineage cells into adherent, motile, mature macrophages. The colony-stimulating factor-1 receptor transduces colony-stimulating factor-1 signaling, and we have shown previously that phosphatidylinositol 3-kinase p110δ is a critical mediator of colony-stimulating factor-1–stimulated motility through the colony-stimulating factor-1 receptor pY721 motif. Src family kinases are also implicated in the regulation of macrophage motility and in colony-stimulating factor-1 receptor signaling, although functional redundancy of the multiple SFKs expressed in macrophages makes it challenging to delineate their specific functions. We report a comprehensive analysis of individual Src family kinase expression in macrophage cell lines and primary macrophages and demonstrate colony-stimulating factor-1–induced changes in Src family kinase subcellular localization, which provides clues to their distinct and redundant functions in macrophages. Moreover, expression of individual Src family kinases is both species specific and dependent on colony-stimulating factor-1–induced macrophage differentiation. Hck associated with the activated colony-stimulating factor-1 receptor, whereas Lyn associated with the receptor in a constitutive manner. Consistent with this, inhibitor studies revealed that Src family kinases were important for both colony-stimulating factor-1 receptor activation and colony-stimulating factor-1–induced macrophage spreading, motility, and invasion. Distinct colony-stimulating factor-1–induced changes in the subcellular localization of individual SFKs suggest specific roles for these Src family kinases in the macrophage response to colony-stimulating factor-1.
- Published
- 2016
- Full Text
- View/download PDF
50. Micro(glial)-managing executive function: white matter inflammation drives catatonia
- Author
-
Sarah E. Pease-Raissi and Jonah R. Chan
- Subjects
Male ,0301 basic medicine ,Medical and Health Sciences ,Mice ,Executive Function ,Myelin ,0302 clinical medicine ,2',3'-Cyclic Nucleotide 3'-Phosphodiesterase ,Prevalence ,2.1 Biological and endogenous factors ,Organic Chemicals ,Aetiology ,Myelin Sheath ,Mice, Knockout ,Microglia ,Age Factors ,Brain ,Phosphodiesterase ,Catatonia ,General Medicine ,Middle Aged ,White Matter ,Oligodendroglia ,Phenotype ,Mental Health ,medicine.anatomical_structure ,Schizophrenia ,Female ,medicine.symptom ,Research Article ,Adult ,Genotype ,Immunology ,Receptor, Macrophage Colony-Stimulating Factor ,Inflammation ,White matter ,03 medical and health sciences ,medicine ,Animals ,Humans ,Alleles ,Neuroinflammation ,business.industry ,Neurosciences ,medicine.disease ,Brain Disorders ,Mice, Inbred C57BL ,030104 developmental biology ,Mutation ,Commentary ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The underlying cellular mechanisms of catatonia, an executive "psychomotor" syndrome that is observed across neuropsychiatric diseases, have remained obscure. In humans and mice, reduced expression of the structural myelin protein CNP is associated with catatonic signs in an age-dependent manner, pointing to the involvement of myelin-producing oligodendrocytes. Here, we showed that the underlying cause of catatonic signs is the low-grade inflammation of white matter tracts, which marks a final common pathway in Cnp-deficient and other mutant mice with minor myelin abnormalities. The inhibitor of CSF1 receptor kinase signaling PLX5622 depleted microglia and alleviated the catatonic symptoms of Cnp mutants. Thus, microglia and low-grade inflammation of myelinated tracts emerged as the trigger of a previously unexplained mental condition. We observed a very high (25%) prevalence of individuals with catatonic signs in a deeply phenotyped schizophrenia sample (n = 1095). Additionally, we found the loss-of-function allele of a myelin-specific gene (CNP rs2070106-AA) associated with catatonia in 2 independent schizophrenia cohorts and also associated with white matter hyperintensities in a general population sample. Since the catatonic syndrome is likely a surrogate marker for other executive function defects, we suggest that microglia-directed therapies may be considered in psychiatric disorders associated with myelin abnormalities.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.