1
|
Hikosaka M, Parvez MSA, Yamawaki Y, Oe S, Liang Y, Wada Y, Hirahara Y, Koike T, Imai H, Oishi N, Schalbetter SM, Kumagai A, Yoshida M, Sakurai T, Kitada M, Meyer U, Narumiya S, Ohtsuki G. Maternal immune activation followed by peripubertal stress combinedly produce reactive microglia and confine cerebellar cognition. Commun Biol 2025; 8:296. [PMID: 40033126 DOI: 10.1038/s42003-025-07566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025] Open
Abstract
The functional alteration of microglia arises in brains exposed to external stress during early development. Pathophysiological findings of neurodevelopmental disorders such as schizophrenia and autism spectrum disorder suggest cerebellar functional deficits. However, the link between stress-induced microglia reactivity and cerebellar dysfunction is missing. Here, we investigate the developmental immune environment in translational mouse models that combine two risk factors: maternal infection and repeated social defeat stress (2HIT). We find the synergy of inflammatory stress insults, leading to microglial increase specifically in the cerebellum of both sexes. Microglial turnover correlates with the Purkinje neuron loss in 2HIT mice. Highly multiplexed imaging-mass-cytometry identifies a cell transition to TREM2(+) stress-associated microglia in the cerebellum. Single-cell-proteomic clustering reveals IL-6- and TGFβ-signaling association with microglial cell transitions. Reduced excitability of remaining Purkinje cells, cerebellum-involved brain-wide functional dysconnectivity, and behavioral abnormalities indicate cerebellar cognitive dysfunctions in 2HIT animals, which are ameliorated by both systemic and cerebellum-specific microglia replacement.
Collapse
Affiliation(s)
- Momoka Hikosaka
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Md Sorwer Alam Parvez
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuki Yamawaki
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Souichi Oe
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Yuan Liang
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
- Institute of Basic Theory in Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yayoi Wada
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Yukie Hirahara
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Taro Koike
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Hirohiko Imai
- Department of Systems Science, Kyoto University Graduate School of Informatics, Yoshida-Honmachi, Kyoto, Japan
- Innovation Research Center for Quantum Medicine, Gifu University School of Medicine, Gifu, Japan
| | - Naoya Oishi
- Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sina M Schalbetter
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | | | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takeshi Sakurai
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Hirakata-shi, Osaka, Japan
| | - Urs Meyer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan
| | - Gen Ohtsuki
- Department of Drug Discovery Medicine, Kyoto University, Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
2
|
Qin Z, Han Y, Du Y, Zhang Y, Bian Y, Wang R, Wang H, Guo F, Yuan H, Pan Y, Jin J, Zhou Q, Wang Y, Han F, Xu Y, Jiang J. Bioactive materials from berberine-treated human bone marrow mesenchymal stem cells promote alveolar bone regeneration by regulating macrophage polarization. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1010-1026. [PMID: 38489007 DOI: 10.1007/s11427-023-2454-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/19/2023] [Indexed: 03/17/2024]
Abstract
Alveolar bone regeneration has been strongly linked to macrophage polarization. M1 macrophages aggravate alveolar bone loss, whereas M2 macrophages reverse this process. Berberine (BBR), a natural alkaloid isolated and refined from Chinese medicinal plants, has shown therapeutic effects in treating metabolic disorders. In this study, we first discovered that culture supernatant (CS) collected from BBR-treated human bone marrow mesenchymal stem cells (HBMSCs) ameliorated periodontal alveolar bone loss. CS from the BBR-treated HBMSCs contained bioactive materials that suppressed the M1 polarization and induced the M2 polarization of macrophages in vivo and in vitro. To clarify the underlying mechanism, the bioactive materials were applied to different animal models. We discovered macrophage colony-stimulating factor (M-CSF), which regulates macrophage polarization and promotes bone formation, a key macromolecule in the CS. Injection of pure M-CSF attenuated experimental periodontal alveolar bone loss in rats. Colony-stimulating factor 1 receptor (CSF1R) inhibitor or anti-human M-CSF (M-CSF neutralizing antibody, Nab) abolished the therapeutic effects of the CS of BBR-treated HBMSCs. Moreover, AKT phosphorylation in macrophages was activated by the CS, and the AKT activator reversed the negative effect of the CSF1R inhibitor or Nab. These results suggest that the CS of BBR-treated HBMSCs modulates macrophage polarization via the M-CSF/AKT axis. Further studies also showed that CS of BBR-treated HBMSCs accelerated bone formation and M2 polarization in rat teeth extraction sockets. Overall, our findings established an essential role of BBR-treated HBMSCs CS and this might be the first report to show that the products of BBR-treated HBMSCs have active effects on alveolar bone regeneration.
Collapse
Affiliation(s)
- Ziyue Qin
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yifei Du
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Yixuan Zhang
- Gusu school, Nanjing medical university, Suzhou, 215002, China
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yifeng Bian
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Ruyu Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Haoran Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Fanyi Guo
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Hua Yuan
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
| | - Yongchu Pan
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jianliang Jin
- Department of Human Anatomy, Research Centre for Bone and Stem Cells, Key Laboratory for Aging & Disease, The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Qigang Zhou
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing Jiangsu, 211166, China
| | - Yuli Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China.
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Yan Xu
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210029, China.
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Jiandong Jiang
- Department of Virology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| |
Collapse
|
3
|
Zhao Y, Jiang Y, Shen Y, Su LD. Sepsis Impairs Purkinje Cell Functions and Motor Behaviors Through Microglia Activation. CEREBELLUM (LONDON, ENGLAND) 2024; 23:329-339. [PMID: 36790600 DOI: 10.1007/s12311-023-01531-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
The most common clinical manifestation of sepsis-related encephalopathy (SAE) is the deterioration of cognitive function. Besides, increasing evidence shows that SAE patients exhibit coordination and sensorimotor dysfunctions, suggesting that SAE affects motor function with unclear mechanism. In the present work, we explored the effects of SAE on cerebellar Purkinje cells (PCs) using cecal ligation and perforation (CLP), a standard model for inducing sepsis symptoms similar to those in human patients. Our results show that the sepsis can activate microglia in the cerebellum and promote the secretion of inflammatory factor TNF-α, which increases intrinsic excitability and synaptic transmission of PCs, inhibits the synaptic plasticity of PCs, and impairs motor learning of mice. These findings address how SAE changes PC functions, and thereby are of great significance to reveal pathophysiological feathers of human patients suffering from SAE.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Physiology and Neuroscience Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yao Jiang
- Department of Physiology and Neuroscience Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ying Shen
- Department of Physiology and Neuroscience Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Li-Da Su
- Department of Physiology and Neuroscience Care Unit, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Key Laboratory of the Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou, 310009, China.
| |
Collapse
|
4
|
van der Wildt B, Klockow JL, Miao Z, Reyes ST, Park JH, Shen B, Chin FT. Discovery of a CSF-1R inhibitor and PET tracer for imaging of microglia and macrophages in the brain. Nucl Med Biol 2022; 114-115:99-107. [PMID: 36371938 DOI: 10.1016/j.nucmedbio.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/06/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
Colony stimulating factor 1 receptor (CSF-1R) is a kinase expressed on macrophages and microglia in the brain. It has been recognized as a potential drug and imaging target in treatment of neuroinflammatory diseases and glioblastoma. Despite several attempts, no validated CSF-1R PET tracer is currently available. The aim of this work was to develop a brain permeable CSF-1R PET tracer for non-invasive imaging of CSF-1R in vivo. Based on fragments of two potent and selective CSF-1R inhibitors, novel hybrid molecules were designed and synthesized. Affinity for human recombinant CSF-1R and selectivity over c-KIT and PDGFR-β was determined using a FRET based in vitro assay. Radiosynthesis was performed by fully automated [11C]CH3I methylation of the corresponding des-methyl precursor. PET imaging was performed at baseline, efflux transporter blocking and CSF-1R blocking conditions. Moreover, tracer distribution and blood and plasma radiometabolites were determined following injection in healthy mice. The most promising CSF-1R inhibitor, compound 4, showed high selectivity and high affinity for CSF-1R (IC50: 12 ± 3 nM) and no affinity for kinase family members c-KIT and PDGFR-beta. [11C]4 was obtained in good yield (15 ± 0.2 % decay corrected yield, (2.0 ± 0.26 GBq at end of synthesis) and excellent purity. The compound demonstrated high brain penetration and good metabolic stability (>2 %ID/g at 60 min post injection and 79 ± 8 % intact [11C]4 in brain at 60 min post injection) and no strong efflux transporter substrate behavior. Blocking CSF-1R prior to imaging with [11C]4 resulted in significant decrease in brain uptake. In conclusion, [11C]4 shows good potential as a novel PET tracer for imaging of CSF-1R in the CNS and future experiments in relevant animal models are warranted.
Collapse
Affiliation(s)
- Berend van der Wildt
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jessica L Klockow
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Zheng Miao
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Samantha T Reyes
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Jun H Park
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Bin Shen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA
| | - Frederick T Chin
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
5
|
Yadav S, Priya A, Borade DR, Agrawal-Rajput R. Macrophage subsets and their role: co-relation with colony-stimulating factor-1 receptor and clinical relevance. Immunol Res 2022; 71:130-152. [PMID: 36266603 PMCID: PMC9589538 DOI: 10.1007/s12026-022-09330-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/14/2022] [Indexed: 01/10/2023]
Abstract
Macrophages are one of the first innate immune cells to reach the site of infection or injury. Diverse functions from the uptake of pathogen or antigen, its killing, and presentation, the release of pro- or anti-inflammatory cytokines, activation of adaptive immune cells, clearing off tissue debris, tissue repair, and maintenance of tissue homeostasis have been attributed to macrophages. Besides tissue-resident macrophages, the circulating macrophages are recruited to different tissues to get activated. These are highly plastic cells, showing a spectrum of phenotypes depending on the stimulus received from their immediate environment. The macrophage differentiation requires colony-stimulating factor-1 (CSF-1) or macrophage colony-stimulating factor (M-CSF), colony-stimulating factor-2 (CSF-2), or granulocyte–macrophage colony-stimulating factor (GM-CSF) and different stimuli activate them to different phenotypes. The richness of tissue macrophages is precisely controlled via the CSF-1 and CSF-1R axis. In this review, we have given an overview of macrophage origin via hematopoiesis/myelopoiesis, different phenotypes associated with macrophages, their clinical significance, and how they are altered in various diseases. We have specifically focused on the function of CSF-1/CSF-1R signaling in deciding macrophage fate and the outcome of aberrant CSF-1R signaling in relation to macrophage phenotype in different diseases. We further extend the review to briefly discuss the possible strategies to manipulate CSF-1R and its signaling with the recent updates.
Collapse
Affiliation(s)
- Shivani Yadav
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India
| | - Astik Priya
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India
| | - Diksha R Borade
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India.
| |
Collapse
|
6
|
Tang Y, Huang S, Chen X, Huang J, Lin Q, Huang L, Wang S, Zhu Q, Xu Y, Zou Y. Design, Synthesis and Biological Evaluation of Novel and Potent Protein Arginine Methyltransferases 5 Inhibitors for Cancer Therapy. Molecules 2022; 27:molecules27196637. [PMID: 36235174 PMCID: PMC9572541 DOI: 10.3390/molecules27196637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Protein arginine methyltransferases 5 (PRMT5) is a clinically promising epigenetic target that is upregulated in a variety of tumors. Currently, there are several PRMT5 inhibitors under preclinical or clinical development, however the established clinical inhibitors show favorable toxicity. Thus, it remains an unmet need to discover novel and structurally diverse PRMT5 inhibitors with characterized therapeutic utility. Herein, a series of tetrahydroisoquinoline (THIQ) derivatives were designed and synthesized as PRMT5 inhibitors using GSK-3326595 as the lead compound. Among them, compound 20 (IC50: 4.2 nM) exhibits more potent PRMT5 inhibitory activity than GSK-3326595 (IC50: 9.2 nM). In addition, compound 20 shows high anti-proliferative effects on MV-4-11 and MDA-MB-468 tumor cells and low cytotoxicity on AML-12 hepatocytes. Furthermore, compound 20 possesses acceptable pharmacokinetic profiles and displays considerable in vivo antitumor efficacy in a MV-4-11 xenograft model. Taken together, compound 20 is an antitumor compound worthy of further study.
Collapse
Affiliation(s)
- Yixuan Tang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shihui Huang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xingxing Chen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Junzhang Huang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qianwen Lin
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Huang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuping Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qihua Zhu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Design and Optimization, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yungen Xu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Design and Optimization, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (Y.X.); (Y.Z.); Tel.: +86-025-86185303 (Y.X.)
| | - Yi Zou
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Correspondence: (Y.X.); (Y.Z.); Tel.: +86-025-86185303 (Y.X.)
| |
Collapse
|
7
|
Chen C, Park B, Ragonnaud E, Bodogai M, Wang X, Zong L, Lee JM, Beerman I, Biragyn A. Cancer co-opts differentiation of B-cell precursors into macrophage-like cells. Nat Commun 2022; 13:5376. [PMID: 36104343 PMCID: PMC9474882 DOI: 10.1038/s41467-022-33117-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 08/31/2022] [Indexed: 11/08/2022] Open
Abstract
We have recently reported that some cancers induce accumulation of bone marrow (BM) B-cell precursors in the spleen to convert them into metastasis-promoting, immunosuppressive B cells. Here, using various murine tumor models and samples from humans with breast and ovarian cancers, we provide evidence that cancers also co-opt differentiation of these B-cell precursors to generate macrophage-like cells (termed B-MF). We link the transdifferentiation to a small subset of CSF1R+ Pax5Low cells within BM pre-B and immature B cells responding to cancer-secreted M-CSF with downregulation of the transcription factor Pax5 via CSF1R signaling. Although the primary source of tumor-associated macrophages is monocytes, B-MFs are phenotypically and functionally distinguishable. Compared to monocyte-derived macrophages, B-MFs more efficiently phagocytize apoptotic cells, suppress proliferation of T cells and induce FoxP3+ regulatory T cells. In mouse tumor models, B-MFs promote shrinkage of the tumor-infiltrating IFNγ+ CD4 T cell pool and increase cancer progression and metastasis, suggesting that this cancer-induced transdifferentiation pathway is functionally relevant and hence could serve as an immunotherapeutic target.
Collapse
Affiliation(s)
- Chen Chen
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Bongsoo Park
- Epigenetics and Stem Cell Aging Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Emeline Ragonnaud
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Xin Wang
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Le Zong
- Epigenetics and Stem Cell Aging Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Aging Unit, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
8
|
In vitro 3D malignant melanoma model for the evaluation of hypericin-loaded oil-in-water microemulsion in photodynamic therapy. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00202-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
9
|
Cao X, Chen J, Li B, Dang J, Zhang W, Zhong X, Wang C, Raoof M, Sun Z, Yu J, Fakih MG, Feng M. Promoting antibody-dependent cellular phagocytosis for effective macrophage-based cancer immunotherapy. SCIENCE ADVANCES 2022; 8:eabl9171. [PMID: 35302839 PMCID: PMC8932662 DOI: 10.1126/sciadv.abl9171] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/26/2022] [Indexed: 05/16/2023]
Abstract
Macrophages are essential in eliciting antibody-dependent cellular phagocytosis (ADCP) of cancer cells. However, a satisfactory anticancer efficacy of ADCP is contingent on early antibody administration, and resistance develops along with cancer progression. Here, we investigate the mechanisms underlying ADCP and demonstrate an effective combinatorial strategy to potentiate its efficacy. We identified paclitaxel as a universal adjuvant that efficiently potentiated ADCP by a variety of anticancer antibodies in multiple cancers. Rather than eliciting cytotoxicity on cancer cells, paclitaxel polarized macrophages toward a state with enhanced phagocytic ability. Paclitaxel-treated macrophages down-regulated cell surface CSF1R whose expression was negatively correlated with patient survival in multiple malignancies. The suppression of CSF1R in macrophages enhanced ADCP of cancer cells, suggesting a role of CSF1R in regulating macrophage phagocytic ability. Together, these findings define a potent strategy for using conventional anticancer drugs to stimulate macrophage phagocytosis and promote the therapeutic efficacy of clinical anticancer antibodies.
Collapse
Affiliation(s)
- Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jing Chen
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Bolei Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jessica Dang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Wencan Zhang
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xiancai Zhong
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Chongkai Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Mustafa Raoof
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
| | - Zuoming Sun
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jianhua Yu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope, Duarte, CA 91010, USA
| | - Marwan G. Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
10
|
Yamawaki Y, Wada Y, Matsui S, Ohtsuki G. Microglia-triggered hypoexcitability plasticity of pyramidal neurons in the rat medial prefrontal cortex. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100028. [DOI: 10.1016/j.crneur.2022.100028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 12/16/2022] Open
|
11
|
Jiang C, Wang ZN, Kang YC, Chen Y, Lu WX, Ren HJ, Hou BR. Ki20227 aggravates apoptosis, inflammatory response, and oxidative stress after focal cerebral ischemia injury. Neural Regen Res 2022; 17:137-143. [PMID: 34100449 PMCID: PMC8451550 DOI: 10.4103/1673-5374.314318] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The survival of microglia depends on the colony-stimulating factor-1 receptor (CSF1R) signaling pathway under physiological conditions. Ki20227 is a highly selective CSF1R inhibitor that has been shown to change the morphology of microglia. However, the effects of Ki20227 on the progression of ischemic stroke are unclear. In this study, male C57BL/6 mouse models of focal cerebral ischemic injury were established through the occlusion of the middle cerebral artery and then administered 3 mg/g Ki20227 for 3 successive days. The results revealed that the number of ionized calcium-binding adaptor molecule 1/bromodeoxyuridine double positive cells in the infarct tissue was reduced, the degree of edema was increased, neurological deficits were aggravated, infarct volume was increased, and the number of peri-infarct Nissl bodies was reduced. The number of terminal deoxynucleotidyl transferase dUTP nick-end labeling-positive cells in the peri-infarct tissue was increased. The expression levels of Bax and Cleaved caspase-3 were up-regulated. Bcl-2 expression was downregulated. The expression levels of inflammatory factors and oxidative stress-associated factors were increased. These findings suggested that Ki20227 blocked microglial proliferation and aggravated the pathological progression of ischemia/reperfusion injury in a transient middle cerebral artery occlusion model. This study was approved by the Animal Ethics Committee of Lanzhou University Second Hospital (approval No. D2020-68) on March 6, 2020.
Collapse
Affiliation(s)
- Cheng Jiang
- Department of Neurosurgery, Lanzhou University Second Hospital; Institute of Neurology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Ze-Ning Wang
- Department of Neurosurgery, Lanzhou University Second Hospital; Institute of Neurology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yu-Chen Kang
- Department of Neurosurgery, Lanzhou University Second Hospital; Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yi Chen
- Department of Neurosurgery, Lanzhou University Second Hospital; Institute of Neurology, Lanzhou University, Lanzhou, Gansu Province, China
| | - Wei-Xin Lu
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Hai-Jun Ren
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| | - Bo-Ru Hou
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, Gansu Province, China
| |
Collapse
|
12
|
Crapser JD, Arreola MA, Tsourmas KI, Green KN. Microglia as hackers of the matrix: sculpting synapses and the extracellular space. Cell Mol Immunol 2021; 18:2472-2488. [PMID: 34413489 PMCID: PMC8546068 DOI: 10.1038/s41423-021-00751-3] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/26/2021] [Indexed: 02/08/2023] Open
Abstract
Microglia shape the synaptic environment in health and disease, but synapses do not exist in a vacuum. Instead, pre- and postsynaptic terminals are surrounded by extracellular matrix (ECM), which together with glia comprise the four elements of the contemporary tetrapartite synapse model. While research in this area is still just beginning, accumulating evidence points toward a novel role for microglia in regulating the ECM during normal brain homeostasis, and such processes may, in turn, become dysfunctional in disease. As it relates to synapses, microglia are reported to modify the perisynaptic matrix, which is the diffuse matrix that surrounds dendritic and axonal terminals, as well as perineuronal nets (PNNs), specialized reticular formations of compact ECM that enwrap neuronal subsets and stabilize proximal synapses. The interconnected relationship between synapses and the ECM in which they are embedded suggests that alterations in one structure necessarily affect the dynamics of the other, and microglia may need to sculpt the matrix to modify the synapses within. Here, we provide an overview of the microglial regulation of synapses, perisynaptic matrix, and PNNs, propose candidate mechanisms by which these structures may be modified, and present the implications of such modifications in normal brain homeostasis and in disease.
Collapse
Affiliation(s)
- Joshua D. Crapser
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Miguel A. Arreola
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kate I. Tsourmas
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| | - Kim N. Green
- grid.266093.80000 0001 0668 7243Department of Neurobiology and Behavior, University of California, Irvine, CA USA
| |
Collapse
|
13
|
Lee KH, Yen WC, Lin WH, Wang PC, Lai YL, Su YC, Chang CY, Wu CS, Huang YC, Yang CM, Chou LH, Yeh TK, Chen CT, Shih C, Hsieh HP. Discovery of BPR1R024, an Orally Active and Selective CSF1R Inhibitor that Exhibits Antitumor and Immunomodulatory Activity in a Murine Colon Tumor Model. J Med Chem 2021; 64:14477-14497. [PMID: 34606263 DOI: 10.1021/acs.jmedchem.1c01006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
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.
Collapse
Affiliation(s)
- Kun-Hung Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
- Department of Chemistry, National Tsing Hua University, Hsinchu City 300, Taiwan, ROC
- Biomedical Translation Research Center, Academia Sinica, Taipei City 115, Taiwan, ROC
| | - Wan-Ching Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Pei-Chen Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - You-Liang Lai
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Yu-Chieh Su
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Chun-Yu Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Cai-Syuan Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
- Department of Chemistry, National Tsing Hua University, Hsinchu City 300, Taiwan, ROC
| | - Yu-Chen Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Chen-Ming Yang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Ling-Hui Chou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350, Taiwan, ROC
- Department of Chemistry, National Tsing Hua University, Hsinchu City 300, Taiwan, ROC
- Biomedical Translation Research Center, Academia Sinica, Taipei City 115, Taiwan, ROC
| |
Collapse
|
14
|
Benn CL, Dawson LA. Clinically Precedented Protein Kinases: Rationale for Their Use in Neurodegenerative Disease. Front Aging Neurosci 2020; 12:242. [PMID: 33117143 PMCID: PMC7494159 DOI: 10.3389/fnagi.2020.00242] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Kinases are an intensively studied drug target class in current pharmacological research as evidenced by the large number of kinase inhibitors being assessed in clinical trials. Kinase-targeted therapies have potential for treatment of a broad array of indications including central nervous system (CNS) disorders. In addition to the many variables which contribute to identification of a successful therapeutic molecule, drug discovery for CNS-related disorders also requires significant consideration of access to the target organ and specifically crossing the blood-brain barrier (BBB). To date, only a small number of kinase inhibitors have been reported that are specifically designed to be BBB permeable, which nonetheless demonstrates the potential for success. This review considers the potential for kinase inhibitors in the context of unmet medical need for neurodegenerative disease. A subset of kinases that have been the focus of clinical investigations over a 10-year period have been identified and discussed individually. For each kinase target, the data underpinning the validity of each in the context of neurodegenerative disease is critically evaluated. Selected molecules for each kinase are identified with information on modality, binding site and CNS penetrance, if known. Current clinical development in neurodegenerative disease are summarized. Collectively, the review indicates that kinase targets with sufficient rationale warrant careful design approaches with an emphasis on improving brain penetrance and selectivity.
Collapse
|
15
|
Denny WA, Flanagan JU. Small-molecule CSF1R kinase inhibitors; review of patents 2015-present. Expert Opin Ther Pat 2020; 31:107-117. [PMID: 33108917 DOI: 10.1080/13543776.2021.1839414] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Colony stimulating factor 1 receptor (CSF-1R, also known as c-FMS kinase) is in the class III receptor tyrosine kinase family, along with c-Kit, Flt3 and PDGFRα. CSF-1/CSF-1R signaling promotes the differentiation and survival of myeloid progenitors into populations of monocytes, macrophages, dendritic cells and osteoclasts, as well as microglial cells and also recruits host macrophages to develop into tumor-associated macrophages (TAMs), which promote tumor progression and metastasis. AREAS COVERED In the last 5 years, and recently stimulated by the approval of pexidartinib (Turalio™, Daiichi Sankyo) in 2019 for the treatment of tenosynovial giant cell tumors, there has been a large increase in activity (both journal articles and patent applications) around small molecule inhibitors of CSF1R. Features of this work have been the surprising diversity of chemical classes shown to be potent and selective inhibitors, and the breadth of disease states (cancer, arthritis, and 'cytokine storm' syndromes) covered by CSF1R inhibitors. All these aspects are covered in the following sections. EXPERT OPINION The field has developed rapidly from 2014 to the present, with many different chemotypes proving to be potent inhibitors. The range of potential utilities of CSF1R inhibitors has also expanded to include dementia, ulcerative colitis/Crohn's disease, rheumatoid arthritis inflammation, and fibrosis.
Collapse
Affiliation(s)
- William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences and Maurice Wilkins Centre, University of Auckland , Auckland, New Zealand
| | - Jack U Flanagan
- Auckland Cancer Society Research Centre, School of Medical Sciences and Maurice Wilkins Centre, University of Auckland , Auckland, New Zealand.,Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, University of Auckland , Auckland, New Zealand
| |
Collapse
|
16
|
Hou B, Jiang C, Wang D, Wang G, Wang Z, Zhu M, Kang Y, Su J, Wei P, Ren H, Ju F. Pharmacological Targeting of CSF1R Inhibits Microglial Proliferation and Aggravates the Progression of Cerebral Ischemic Pathology. Front Cell Neurosci 2020; 14:267. [PMID: 33177990 PMCID: PMC7596178 DOI: 10.3389/fncel.2020.00267] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Ischemic stroke can induce rapid activation of the microglia. It has been reported that the microglia’s survival is dependent on colony-stimulating factor 1 receptor (CSF1R) signaling and that pharmacological inhibition of CSF1R leads to morphological changes in the microglia in the healthy brain. However, the impact of CSF1R inhibition on neuronal structures and motor ability after ischemia–reperfusion remains unclear. In this study, we investigated microglial de-ramification, proliferation, and activation after inhibition of CSF1R by a tyrosine kinase inhibitor (ki20227) in a mouse model of global cerebral ischemia induced by bilateral common carotid artery ligation (BCAL). In addition to microglial morphology, we evaluated the mRNA expression of cytokines, chemokines, and inflammatory receptors. Our results show that pharmacological inhibition of CSF1R in ischemic mice resulted in the blockade of microglial proliferation and a shift in microglial morphology reflected by excessive de-ramification and a more activated phenotype accompanied by an enhanced innate immune response. Furthermore, we show that pharmacological inhibition of CSF1R in ischemic mice resulted in the aggravation of neuronal degeneration and behavioral impairment. Intravital two-photon imaging revealed that although pharmacological inhibition of CSF1R did not affect the recovery of dendritic structures, it caused a significant increase in spine elimination during reperfusion in ischemic mice. These findings suggest that pharmacological inhibition of CSF1R induces a blockade of microglial proliferation and causes acute activation of the microglia accompanied by a severe inflammatory response. It aggravates neuronal degeneration, loss of dendritic spines, and behavioral deficits after transient global cerebral ischemia.
Collapse
Affiliation(s)
- Boru Hou
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Cheng Jiang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Dong Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Gang Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Zening Wang
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Miaojuan Zhu
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuchen Kang
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiacheng Su
- Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pengfei Wei
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haijun Ren
- Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Furong Ju
- Shenzhen Key Lab of Neuropsychiatric Modulation and Collaborative Innovation Center for Brain Science, CAS Center for Excellence in Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
17
|
Yamamoto M, Kim M, Imai H, Itakura Y, Ohtsuki G. Microglia-Triggered Plasticity of Intrinsic Excitability Modulates Psychomotor Behaviors in Acute Cerebellar Inflammation. Cell Rep 2020; 28:2923-2938.e8. [PMID: 31509752 DOI: 10.1016/j.celrep.2019.07.078] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 04/20/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cerebellar dysfunction relates to various psychiatric disorders, including autism spectrum and depressive disorders. However, the physiological aspect is less advanced. Here, we investigate the immune-triggered hyperexcitability in the cerebellum on a wider scope. Activated microglia via exposure to bacterial endotoxin lipopolysaccharide or heat-killed Gram-negative bacteria induce a potentiation of the intrinsic excitability in Purkinje neurons, which is suppressed by microglia-activity inhibitor and microglia depletion. An inflammatory cytokine, tumor necrosis factor alpha (TNF-α), released from microglia via toll-like receptor 4, triggers this plasticity. Our two-photon FRET ATP imaging shows an increase in ATP concentration following endotoxin exposure. Both TNF-α and ATP secretion facilitate synaptic transmission. Region-specific inflammation in the cerebellum in vivo shows depression- and autistic-like behaviors. Furthermore, both TNF-α inhibition and microglia depletion revert such behavioral abnormality. Resting-state functional MRI reveals overconnectivity between the inflamed cerebellum and the prefrontal neocortical regions. Thus, immune activity in the cerebellum induces neuronal hyperexcitability and disruption of psychomotor behaviors in animals.
Collapse
Affiliation(s)
- Masamichi Yamamoto
- Department of Nephrology, Kyoto University Graduate School of Medicine, Kyoto University Hospital, Shogoin-Kawaramachi-cho, Sakyo-ward, Kyoto 606-8507, Japan
| | - Minsoo Kim
- The Hakubi Center for Advanced Research, Kyoto University, Yoshida, Sakyo-ward, Kyoto 606-8501, Japan; Department of Molecular and Cellular Physiology, Kyoto University Graduate School of Medicine, Yoshida-Konoe-cho, Sakyo-ward, Kyoto 606-8501, Japan
| | - Hirohiko Imai
- Department of Systems Science, Kyoto University Graduate School of Informatics, Yoshida-Honmachi, Sakyo-ward, Kyoto 606-8501, Japan
| | - Yamato Itakura
- Department of Biophysics, Kyoto University Graduate School of Science, Kitashirakawa-Oiwake-cho, Sakyo-ward, Kyoto 606-8502, Japan
| | - Gen Ohtsuki
- The Hakubi Center for Advanced Research, Kyoto University, Yoshida, Sakyo-ward, Kyoto 606-8501, Japan; Department of Biophysics, Kyoto University Graduate School of Science, Kitashirakawa-Oiwake-cho, Sakyo-ward, Kyoto 606-8502, Japan.
| |
Collapse
|
18
|
Green KN, Crapser JD, Hohsfield LA. To Kill a Microglia: A Case for CSF1R Inhibitors. Trends Immunol 2020; 41:771-784. [PMID: 32792173 PMCID: PMC7484341 DOI: 10.1016/j.it.2020.07.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/02/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Microglia, the brain's immune sentinels, have garnered much attention in recent years. Researchers have begun to identify the manifold roles that these cells play in the central nervous system (CNS), and this work has been greatly facilitated by microglial depletion paradigms. The varying degrees of spatiotemporal manipulation afforded by such techniques allow microglial ablation before, during, and/or following insult, injury, or disease. We review the major methods of microglial depletion, including toxin-based, genetic, and pharmacological approaches, which differ in key factors including depletion onset, duration, and off-target effects. We conclude that pharmacological CSF1R inhibitors afford the most extensive versatility in manipulating microglia, making them ideal candidates for future studies investigating microglial function in health and disease.
Collapse
Affiliation(s)
- Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA.
| | - Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| |
Collapse
|
19
|
Xun Q, Wang Z, Hu X, Ding K, Lu X. Small-Molecule CSF1R Inhibitors as Anticancer Agents. Curr Med Chem 2020; 27:3944-3966. [PMID: 31215373 DOI: 10.2174/1573394715666190618121649] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/20/2019] [Accepted: 05/30/2019] [Indexed: 12/20/2022]
Abstract
Persuasive evidence has been presented linking the infiltration of Tumor-Associated Macrophages (TAMs) with the driving force of tumorigenesis and in the suppression of antitumor immunity. In this context CSF1R, the cellular receptor for Colony Stimulating Factor-1 (CSF1) and Interleukin 34 (IL-34), occupies a central role in manipulating the behavior of TAMs and the dysregulation of CSF1R signaling has been implicated in cancer progression and immunosuppression in many specific cancers. Consequently, CSF1R kinase has been a target of great interest in cancer treatment and significant research efforts have focused on the development of smallmolecule CSF1R inhibitors. In this review, we highlight current progress on the development of these small molecule CSF1R inhibitors as anticancer agents. Special attention is paid to the compounds available in advanced clinical trials.
Collapse
Affiliation(s)
- Qiuju Xun
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Zhen Wang
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xianglong Hu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Ke Ding
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Xiaoyun Lu
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
20
|
Hashimoto A, Fukumoto T, Zhang R, Gabrilovich D. Selective targeting of different populations of myeloid-derived suppressor cells by histone deacetylase inhibitors. Cancer Immunol Immunother 2020; 69:1929-1936. [PMID: 32435850 DOI: 10.1007/s00262-020-02588-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are widely implicated in negative regulation of immune responses in cancer. Inhibition of class I histone deacetylases (HDAC) with entinostat has anti-MDSC activity. However, as single agent, it did not delay tumor growth in EL4 and LLC tumor models. Here, we found that entinostat reduced immune suppressive activity of only one type of MDSC-polymorphonuclear, PMN-MDSC, whereas it had no effect on monocytic M-MDSC or macrophages. M-MDSC had high amount of class II HDAC-HDAC6, which was further increased after the treatment of mice with entinostat. Inhibition of HDAC6 with ricolinostat reduced suppressive activity of M-MDSC, but did not affect PMN-MDSC or delayed tumor growth. However, combination of entinostat and ricolinostat abrogated suppressive activity of both populations of MDSC and substantially delayed tumor progression. Thus, inactivation of MDSC required targeting of both major subsets of these cells via inhibitors of class I and class II HDAC.
Collapse
Affiliation(s)
- Ayumi Hashimoto
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Rm. 118, 3601 Spruce Str., Philadelphia, PA, 19104, USA
| | - Takeshi Fukumoto
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Rugang Zhang
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Dmitry Gabrilovich
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Rm. 118, 3601 Spruce Str., Philadelphia, PA, 19104, USA.
| |
Collapse
|
21
|
Noyori O, Komohara Y, Nasser H, Hiyoshi M, Ma C, Pan C, Carreras J, Nakamura N, Sato A, Ando K, Okuno Y, Nosaka K, Matsuoka M, Suzu S. Expression of IL-34 correlates with macrophage infiltration and prognosis of diffuse large B-cell lymphoma. Clin Transl Immunology 2019; 8:e1074. [PMID: 31417675 PMCID: PMC6691654 DOI: 10.1002/cti2.1074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
Objectives Infiltration of macrophages through the tyrosine kinase receptor CSF1R is a poor prognosis factor in various solid tumors. Indeed, these tumors produce CSF1R ligand, macrophage colony‐stimulating factor (M‐CSF) or interleukin‐34 (IL‐34). However, the significance of these cytokines, particularly, the newly discovered IL‐34 in haematological malignancies, is not fully understood. We therefore analysed the role of IL‐34 in diffuse large B‐cell lymphoma (DLBCL), the most common subtype of malignant lymphoma. Methods We analysed formalin‐fixed paraffin‐embedded lymphoma tissues of 135 DLBCL patients for the expression of IL‐34 and the number of macrophages, and the survival of these patients. The expression of IL‐34 in DLBCL cell lines and the activity of IL‐34 to induce the migration of monocytic cells were also characterised. Results Several lymphoma tissues showed a clear IL‐34 signal, and such signal was detectable in 36% of patients. DLBCL cell lines also expressed IL‐34. Interestingly, the percentage of IL‐34+ patients in the activated B‐cell subtype was significantly higher than that in the germinal centre B‐cell subtype. More interestingly, IL‐34+ patients showed shorter survival periods and higher number of macrophages in lymphoma tissues. The recruitment of monocytes is likely the first step for the higher macrophage density in the IL‐34+ lymphoma tissues. Indeed, IL‐34 induced the migration of monocytic cells. Conclusion Our results raise the possibility that IL‐34 in lymphoma tissues of DLBCL patients recruits monocytes, leading to the higher number of macrophages in the tissues and poor prognosis of patients. IL‐34 may be an additional therapeutic target of DLBCL.
Collapse
Affiliation(s)
- Osamu Noyori
- International Research Center for Medical Sciences Joint Research Center for Human Retrovirus Infection Kumamoto University Kumamoto Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Hesham Nasser
- International Research Center for Medical Sciences Joint Research Center for Human Retrovirus Infection Kumamoto University Kumamoto Japan
| | - Masateru Hiyoshi
- International Research Center for Medical Sciences Joint Research Center for Human Retrovirus Infection Kumamoto University Kumamoto Japan.,Present address: Department of Safety Research on Blood and Biologics National Institute of Infectious Diseases Tokyo Japan
| | - Chaoya Ma
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Cheng Pan
- Department of Cell Pathology Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Joaquim Carreras
- Department of Pathology School of Medicine Tokai University Kanagawa Japan
| | - Naoya Nakamura
- Department of Pathology School of Medicine Tokai University Kanagawa Japan
| | - Ai Sato
- Department of Hematology and Oncology School of Medicine Tokai University Kanagawa Japan
| | - Kiyoshi Ando
- Department of Hematology and Oncology School of Medicine Tokai University Kanagawa Japan
| | - Yutaka Okuno
- Department of Hematology, Rheumatology, and Infectious Diseases Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Kisato Nosaka
- Department of Hematology, Rheumatology, and Infectious Diseases Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Masao Matsuoka
- Department of Hematology, Rheumatology, and Infectious Diseases Graduate School of Medical Sciences Kumamoto University Kumamoto Japan
| | - Shinya Suzu
- International Research Center for Medical Sciences Joint Research Center for Human Retrovirus Infection Kumamoto University Kumamoto Japan
| |
Collapse
|
22
|
Liu YQ, Wang YN, Lu XY, Tong LJ, Li Y, Zhang T, Xun QJ, Feng F, Chen YZ, Su Y, Shen YY, Chen Y, Geng MY, Ding K, Li YL, Xie H, Ding J. Identification of compound D2923 as a novel anti-tumor agent targeting CSF1R. Acta Pharmacol Sin 2018; 39:1768-1776. [PMID: 29968849 PMCID: PMC6289367 DOI: 10.1038/s41401-018-0056-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 01/08/2023]
Abstract
Colony-stimulating factor 1 receptor (CSF1R) plays a critical role in promoting tumor progression in various types of tumors. Here, we identified D2923 as a novel and selective inhibitor of CSF1R and explored its antitumor activity both in vitro and in vivo. D2923 potently inhibited CSF1R in vitro kinase activity with an IC50 value of 0.3 nM. It exhibited 10- to 300-fold less potency against a panel of kinases tested. D2923 markedly blocked CSF-1-induced activation of CSF1R and its downstream signaling transduction in THP-1 and RAW264.7 macrophages and thus inhibited the in vitro growth of macrophages. Moreover, D2923 dose-dependently attenuated the proliferation of a small panel of myeloid leukemia cells, mainly by arresting the cells at G1 phase as well as inducing apoptosis in the cells. The results of the in vivo experiments further demonstrated that D2923 displayed potent antitumor activity against M-NFS-60 xenografts, with tumor growth inhibition rates of 50% and 88% at doses of 40 and 80 mg/kg, respectively. Additionally, D2923 was well tolerated with no significant body-weight loss observed in the treatment groups compared with the control. Furthermore, a western blot analysis and the immunohistochemistry results confirmed that the phosphorylation of CSF1R in tumor tissue was dramatically reduced after D2923 treatment, and this was accompanied by the depletion of macrophages in the tumor. Meanwhile, the expression of the proliferation marker Ki67 was also markedly decreased in the D2923 treatment group compared with the control group. Taken together, we identified D2923 as a novel and effective CSF1R inhibitor, which deserves further investigation.
Collapse
Affiliation(s)
- Ying-Qiang Liu
- School of Life Science, Shanghai University, 200444, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ya-Nan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, 100049, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Xiao-Yun Lu
- School of Pharmacy, Jinan University, No. 601 Huangpu Avenue, 510632, West Guangzhou, China
| | - Lin-Jiang Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Yan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Tao Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, 100049, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Qiu-Ju Xun
- School of Pharmacy, University of Chinese Academy of Sciences, 100049, Beijing, China
- School of Pharmacy, Jinan University, No. 601 Huangpu Avenue, 510632, West Guangzhou, China
| | - Fang Feng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Yu-Zhe Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
- School of Pharmacy, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yi Su
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Yan-Yan Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Yi Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Mei-Yu Geng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Ke Ding
- School of Pharmacy, Jinan University, No. 601 Huangpu Avenue, 510632, West Guangzhou, China
| | - Yan-Li Li
- School of Life Science, Shanghai University, 200444, Shanghai, China.
| | - Hua Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.
| | - Jian Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.
| |
Collapse
|
23
|
Abstract
Bone metastasis, or the development of secondary tumors within the bone of cancer patients, is a debilitating and incurable disease. Despite its morbidity, the biology of bone metastasis represents one of the most complex and intriguing of all oncogenic processes. This complexity derives from the intricately organized bone microenvironment in which the various stages of hematopoiesis, osteogenesis, and osteolysis are jointly regulated but spatially restricted. Disseminated tumor cells (DTCs) from various common malignancies such as breast, prostate, lung, and kidney cancers or myeloma are uniquely primed to subvert these endogenous bone stromal elements to grow into pathological osteolytic or osteoblastic lesions. This colonization process can be separated into three key steps: seeding, dormancy, and outgrowth. Targeting the processes of dormancy and initial outgrowth offers the most therapeutic promise. Here, we discuss the concepts of the bone metastasis niche, from controlling tumor-cell survival to growth into clinically detectable disease.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| | - Theresa Guise
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544
| |
Collapse
|
24
|
Xun Q, Zhang Z, Luo J, Tong L, Huang M, Wang Z, Zou J, Liu Y, Xu Y, Xie H, Tu ZC, Lu X, Ding K. Design, Synthesis, and Structure–Activity Relationship Study of 2-Oxo-3,4-dihydropyrimido[4,5-d]pyrimidines as New Colony Stimulating Factor 1 Receptor (CSF1R) Kinase Inhibitors. J Med Chem 2018; 61:2353-2371. [PMID: 29499108 DOI: 10.1021/acs.jmedchem.7b01612] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Qiuju Xun
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Zhang Zhang
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Jinfeng Luo
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Linjiang Tong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Minhao Huang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Zhen Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Jian Zou
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Yingqiang Liu
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
- School of Life Sciences, Shanghai University, No. 99 Shangda Road, Shanghai 200444, China
| | - Yong Xu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Hua Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Zheng-Chao Tu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Xiaoyun Lu
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Ke Ding
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- School of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
- Guangzhou City Key Laboratory of Precision Chemical Drug Development, Guangzhou 510632, China
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MOE) of People’s Republic of China, Guangzhou 510632, China
| |
Collapse
|
25
|
Chia K, Mazzolini J, Mione M, Sieger D. Tumor initiating cells induce Cxcr4-mediated infiltration of pro-tumoral macrophages into the brain. eLife 2018; 7:e31918. [PMID: 29465400 PMCID: PMC5821457 DOI: 10.7554/elife.31918] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/31/2018] [Indexed: 12/28/2022] Open
Abstract
It is now clear that microglia and macrophages are present in brain tumors, but whether or how they affect initiation and development of tumors is not known. Exploiting the advantages of the zebrafish (Danio rerio) model, we showed that macrophages and microglia respond immediately upon oncogene activation in the brain. Overexpression of human AKT1 within neural cells of larval zebrafish led to a significant increase in the macrophage and microglia populations. By using a combination of transgenic and mutant zebrafish lines, we showed that this increase was caused by the infiltration of peripheral macrophages into the brain mediated via Sdf1b-Cxcr4b signaling. Intriguingly, confocal live imaging reveals highly dynamic interactions between macrophages/microglia and pre-neoplastic cells, which do not result in phagocytosis of pre-neoplastic cells. Finally, depletion of macrophages and microglia resulted in a significant reduction of oncogenic cell proliferation. Thus, macrophages and microglia show tumor promoting functions already during the earliest stages of the developing tumor microenvironment.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Marina Mione
- Centre for Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
26
|
Kumar V, Donthireddy L, Marvel D, Condamine T, Wang F, Lavilla-Alonso S, Hashimoto A, Vonteddu P, Behera R, Goins MA, Mulligan C, Nam B, Hockstein N, Denstman F, Shakamuri S, Speicher DW, Weeraratna AT, Chao T, Vonderheide RH, Languino LR, Ordentlich P, Liu Q, Xu X, Lo A, Puré E, Zhang C, Loboda A, Sepulveda MA, Snyder LA, Gabrilovich DI. Cancer-Associated Fibroblasts Neutralize the Anti-tumor Effect of CSF1 Receptor Blockade by Inducing PMN-MDSC Infiltration of Tumors. Cancer Cell 2017; 32:654-668.e5. [PMID: 29136508 PMCID: PMC5827952 DOI: 10.1016/j.ccell.2017.10.005] [Citation(s) in RCA: 476] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/25/2017] [Accepted: 10/09/2017] [Indexed: 02/07/2023]
Abstract
Tumor-associated macrophages (TAM) contribute to all aspects of tumor progression. Use of CSF1R inhibitors to target TAM is therapeutically appealing, but has had very limited anti-tumor effects. Here, we have identified the mechanism that limited the effect of CSF1R targeted therapy. We demonstrated that carcinoma-associated fibroblasts (CAF) are major sources of chemokines that recruit granulocytes to tumors. CSF1 produced by tumor cells caused HDAC2-mediated downregulation of granulocyte-specific chemokine expression in CAF, which limited migration of these cells to tumors. Treatment with CSF1R inhibitors disrupted this crosstalk and triggered a profound increase in granulocyte recruitment to tumors. Combining CSF1R inhibitor with a CXCR2 antagonist blocked granulocyte infiltration of tumors and showed strong anti-tumor effects.
Collapse
Affiliation(s)
- Vinit Kumar
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Douglas Marvel
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Thomas Condamine
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Fang Wang
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Sergio Lavilla-Alonso
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ayumi Hashimoto
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Prashanthi Vonteddu
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Reeti Behera
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Marlee A Goins
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Charles Mulligan
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Brian Nam
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Neil Hockstein
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Fred Denstman
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - Shanti Shakamuri
- Helen F. Graham Cancer Center at Christiana Care Health System, Wilmington, DE, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ashani T Weeraratna
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Timothy Chao
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | - Lucia R Languino
- Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Qin Liu
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Albert Lo
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Ellen Puré
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Chunsheng Zhang
- Department of Genetics and Pharmacogenomics, MRL, Merck & Co., Inc., Boston, MA 02115, USA
| | - Andrey Loboda
- Department of Genetics and Pharmacogenomics, MRL, Merck & Co., Inc., Boston, MA 02115, USA
| | | | | | - Dmitry I Gabrilovich
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Wang W, Wang H, Zhou X, Li X, Sun W, Dellinger M, Boyce BF, Xing L. Lymphatic Endothelial Cells Produce M-CSF, Causing Massive Bone Loss in Mice. J Bone Miner Res 2017; 32:939-950. [PMID: 28052488 PMCID: PMC5413433 DOI: 10.1002/jbmr.3077] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/22/2016] [Accepted: 12/30/2016] [Indexed: 11/08/2022]
Abstract
Gorham-Stout disease (GSD) is a rare bone disorder characterized by aggressive osteolysis associated with lymphatic vessel invasion within bone marrow cavities. The etiology of GSD is not known, and there is no effective therapy or animal model for the disease. Here, we investigated if lymphatic endothelial cells (LECs) affect osteoclasts (OCs) to cause a GSD osteolytic phenotype in mice. We examined the effect of a mouse LEC line on osteoclastogenesis in co-cultures. LECs significantly increased receptor activator of NF-κB ligand (RANKL)-mediated OC formation and bone resorption. LECs expressed high levels of macrophage colony-stimulating factor (M-CSF), but not RANKL, interleukin-6 (IL-6), and tumor necrosis factor (TNF). LEC-mediated OC formation and bone resorption were blocked by an M-CSF neutralizing antibody or Ki20227, an inhibitor of the M-CSF receptor, c-Fms. We injected LECs into the tibias of wild-type (WT) mice and observed massive osteolysis on X-ray and micro-CT scans. Histology showed that LEC-injected tibias had significant trabecular and cortical bone loss and increased OC numbers. M-CSF protein levels were significantly higher in serum and bone marrow plasma of mice given intra-tibial LEC injections. Immunofluorescence staining showed extensive replacement of bone and marrow by podoplanin+ LECs. Treatment of LEC-injected mice with Ki20227 significantly decreased tibial bone destruction. In addition, lymphatic vessels in a GSD bone sample were stained positively for M-CSF. Thus, LECs cause bone destruction in vivo in mice by secreting M-CSF, which promotes OC formation and activation. Blocking M-CSF signaling may represent a new therapeutic approach for treatment of patients with GSD. Furthermore, tibial injection of LECs is a useful mouse model to study GSD. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Wensheng Wang
- The 1st Affiliated Hospital, Xinxiang Medical University, Xinxiang, China.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hua Wang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Institute of Stomatology, Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Xichao Zhou
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Xing Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Wen Sun
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michael Dellinger
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
28
|
Ihn HJ, Lee D, Lee T, Kim SH, Shin HI, Bae YC, Hong JM, Park EK. Inhibitory Effects of KP-A159, a Thiazolopyridine Derivative, on Osteoclast Differentiation, Function, and Inflammatory Bone Loss via Suppression of RANKL-Induced MAP Kinase Signaling Pathway. PLoS One 2015; 10:e0142201. [PMID: 26536233 PMCID: PMC4633183 DOI: 10.1371/journal.pone.0142201] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 10/19/2015] [Indexed: 11/19/2022] Open
Abstract
Abnormally elevated formation and activation of osteoclasts are primary causes for a majority of skeletal diseases. In this study, we found that KP-A159, a newly synthesized thiazolopyridine derivative, inhibited osteoclast differentiation and function in vitro, and inflammatory bone loss in vivo. KP-A159 did not cause a cytotoxic response in bone marrow macrophages (BMMs), but significantly inhibited the formation of multinucleated tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts induced by macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL). KP-A159 also dramatically inhibited the expression of marker genes related to osteoclast differentiation, including TRAP (Acp5), cathepsin K (Ctsk), dendritic cell-specific transmembrane protein (Dcstamp), matrix metallopeptidase 9 (Mmp9), and nuclear factor of activated T-cells, cytoplasmic 1 (Nfatc1). Moreover, actin ring and resorption pit formation were inhibited by KP-A159. Analysis of the signaling pathway involved showed that KP-A159 inhibited RANKL-induced activation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and mitogen-activated protein kinase kinase1/2 (MEK1/2). In a mouse inflammatory bone loss model, KP-A159 significantly rescued lipopolysaccharide (LPS)-induced bone loss by suppressing osteoclast numbers. Therefore, KP-A159 targets osteoclasts, and may be a potential candidate compound for prevention and/or treatment of inflammatory bone loss.
Collapse
Affiliation(s)
- Hye Jung Ihn
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Doohyun Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Taeho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sang-Hyun Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hong-In Shin
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu, Republic of Korea
| | - Yong Chul Bae
- Department of Oral Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Jung Min Hong
- Skeletal Diseases Genome Research Centre, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
29
|
Baladi T, Abet V, Piguel S. State-of-the-art of small molecule inhibitors of the TAM family: the point of view of the chemist. Eur J Med Chem 2015; 105:220-37. [PMID: 26498569 DOI: 10.1016/j.ejmech.2015.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 10/01/2015] [Accepted: 10/03/2015] [Indexed: 01/04/2023]
Abstract
The TAM family of tyrosine kinases receptors (Tyro3, Axl and Mer) is implicated in cancer development, autoimmune reactions and viral infection and is therefore emerging as an effective and attractive therapeutic target. To date, only a few small molecules have been intentionally designed to block the TAM kinases, while most of the inhibitors were developed for blocking different protein kinases and then identified through selectivity profile studies. This minireview will examine in terms of chemical structure the different compounds able to act on either one, two or three TAM kinases with details about structure-activity relationships, drug-metabolism and pharmacokinetics properties where they exist.
Collapse
Affiliation(s)
- Tom Baladi
- Institut Curie/UMR9187-U1196, 91405 Orsay cedex, France; Univ Paris Sud, 91405 Orsay cedex, France
| | | | - Sandrine Piguel
- Institut Curie/UMR9187-U1196, 91405 Orsay cedex, France; Univ Paris Sud, 91405 Orsay cedex, France.
| |
Collapse
|
30
|
CSF-1/CSF-1R targeting agents in clinical development for cancer therapy. Curr Opin Pharmacol 2015; 23:45-51. [PMID: 26051995 DOI: 10.1016/j.coph.2015.05.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/07/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
Macrophage infiltration has been identified as an independent poor prognostic factor for several cancer entities. In mouse tumor models macrophages orchestrate various tumor-promoting processes. This observation sparked an interest to therapeutically target these plastic innate immune cells. To date, blockade of colony stimulating factor-1 or its receptor represents the only truly selective approach to manipulate macrophages in cancer patients. Here, we discuss the currently available information on efficacy and safety of various CSF-1/CSF-1R inhibitors in cancer patients and highlight potential combination partners emerging from preclinical studies while considering the differences between mouse and human macrophage biology.
Collapse
|
31
|
Li HS, Watowich SS. Innate immune regulation by STAT-mediated transcriptional mechanisms. Immunol Rev 2015; 261:84-101. [PMID: 25123278 DOI: 10.1111/imr.12198] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term innate immunity typically refers to a quick but non-specific host defense response against invading pathogens. The innate immune system comprises particular immune cell populations, epithelial barriers, and numerous secretory mediators including cytokines, chemokines, and defense peptides. Innate immune cells are also now recognized to play important contributing roles in cancer and pathological inflammatory conditions. Innate immunity relies on rapid signal transduction elicited upon pathogen recognition via pattern recognition receptors (PRRs) and cell:cell communication conducted by soluble mediators, including cytokines. A majority of cytokines involved in innate immune signaling use a molecular cascade encompassing receptor-associated Jak protein tyrosine kinases and STAT (signal transducer and activator of transcription) transcriptional regulators. Here, we focus on roles for STAT proteins in three major innate immune subsets: neutrophils, macrophages, and dendritic cells (DCs). While knowledge in this area is only now emerging, understanding the molecular regulation of these cell types is necessary for developing new approaches to treat human disorders such as inflammatory conditions, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Haiyan S Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
32
|
Aikawa Y, Yamagata K, Katsumoto T, Shima Y, Shino M, Stanley ER, Cleary ML, Akashi K, Tenen DG, Kitabayashi I. Essential role of PU.1 in maintenance of mixed lineage leukemia-associated leukemic stem cells. Cancer Sci 2015; 106:227-36. [PMID: 25529853 PMCID: PMC4373983 DOI: 10.1111/cas.12593] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/05/2014] [Accepted: 12/14/2014] [Indexed: 01/24/2023] Open
Abstract
Acute myeloid leukemia is a clonal malignant disorder derived from a small number of leukemic stem cells (LSCs). Rearrangements of the mixed lineage leukemia (MLL) gene are found in acute myeloid leukemia associated with poor prognosis. The upregulation of Hox genes is critical for LSC induction and maintenance, but is unlikely to support malignancy and the high LSC frequency observed in MLL leukemias. The present study shows that MLL fusion proteins interact with the transcription factor PU.1 to activate the transcription of CSF-1R, which is critical for LSC activity. Acute myeloid leukemia is cured by either deletion of PU.1 or ablation of cells expressing CSF-1R. Kinase inhibitors specific for CSF-1R prolong survival time. These findings indicate that PU.1-mediated upregulation of CSF-1R is a critical effector of MLL leukemogenesis.
Collapse
Affiliation(s)
- Yukiko Aikawa
- Division of Hematological Malignancy, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Laoui D, Van Overmeire E, De Baetselier P, Van Ginderachter JA, Raes G. Functional Relationship between Tumor-Associated Macrophages and Macrophage Colony-Stimulating Factor as Contributors to Cancer Progression. Front Immunol 2014; 5:489. [PMID: 25339957 PMCID: PMC4188035 DOI: 10.3389/fimmu.2014.00489] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/22/2014] [Indexed: 12/14/2022] Open
Abstract
The current review article describes the functional relationship between tumor-associated macrophages (TAM) as key cellular contributors to cancer malignancy on the one hand and macrophage-colony-stimulating factor (M-CSF or CSF-1) as an important molecular contributor on the other. We recapitulate the available data on expression of M-CSF and the M-CSF receptor (M-CSFR) in human tumor tissue as constituents of a stromal macrophage signature and on the limits of the predictive and prognostic value of plasma M-CSF levels. After providing an update on current insights into the nature of TAM heterogeneity at the level of M1/M2 phenotype and TAM subsets, we give an overview of experimental evidence, based on genetic, antibody-mediated, and pharmacological disruption of M-CSF/M-CSFR signaling, for the extent to which M-CSFR signaling can not only determine the TAM quantity, but can also contribute to shaping the phenotype and heterogeneity of TAM and other related tumor-infiltrating myeloid cells (TIM). Finally, we review the accumulating information on the – sometimes conflicting – effects blocking M-CSFR signaling may have on various aspects of cancer progression such as tumor growth, invasion, angiogenesis, metastasis, and resistance to therapy and we thereby discuss in how far these different effects actually reflect a contribution of TAM.
Collapse
Affiliation(s)
- Damya Laoui
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Eva Van Overmeire
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| | - Geert Raes
- Myeloid Cell Immunology Laboratory, VIB , Brussels , Belgium ; Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel , Brussels , Belgium
| |
Collapse
|
34
|
Sauter KA, Pridans C, Sehgal A, Tsai YT, Bradford BM, Raza S, Moffat L, Gow DJ, Beard PM, Mabbott NA, Smith LB, Hume DA. Pleiotropic effects of extended blockade of CSF1R signaling in adult mice. J Leukoc Biol 2014; 96:265-74. [PMID: 24652541 PMCID: PMC4378363 DOI: 10.1189/jlb.2a0114-006r] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We investigated the role of CSF1R signaling in adult mice using prolonged treatment with anti-CSF1R antibody. Mutation of the CSF1 gene in the op/op mouse produces numerous developmental abnormalities. Mutation of the CSF1R has an even more penetrant phenotype, including perinatal lethality, because of the existence of a second ligand, IL-34. These effects on development provide limited insight into functions of CSF1R signaling in adult homeostasis. The carcass weight and weight of several organs (spleen, kidney, and liver) were reduced in the treated mice, but overall body weight gain was increased. Despite the complete loss of Kupffer cells, there was no effect on liver gene expression. The treatment ablated OCL, increased bone density and trabecular volume, and prevented the decline in bone mass seen in female mice with age. The op/op mouse has a deficiency in pancreatic β cells and in Paneth cells in the gut wall. Only the latter was reproduced by the antibody treatment and was associated with increased goblet cell number but no change in villus architecture. Male op/op mice are infertile as a result of testosterone insufficiency. Anti-CSF1R treatment ablated interstitial macrophages in the testis, but there was no sustained effect on testosterone or LH. The results indicate an ongoing requirement for CSF1R signaling in macrophage and OCL homeostasis but indicate that most effects of CSF1 and CSF1R mutations are due to effects on development.
Collapse
Affiliation(s)
- Kristin A. Sauter
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Clare Pridans
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Anuj Sehgal
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Yi Ting Tsai
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Scotland, United Kingdom
| | - Barry M. Bradford
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Sobia Raza
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Lindsey Moffat
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Deborah J. Gow
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Philippa M. Beard
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Neil A. Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and
| | - Lee B. Smith
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Scotland, United Kingdom
| | - David A. Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies and ,Correspondence: The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Scotland EH25 9RG, UK. E-mail:
| |
Collapse
|
35
|
Mainetti LE, Zhe X, Diedrich J, Saliganan AD, Cho WJ, Cher ML, Heath E, Fridman R, Kim HRC, Bonfil RD. Bone-induced c-kit expression in prostate cancer: a driver of intraosseous tumor growth. Int J Cancer 2014; 136:11-20. [PMID: 24798488 DOI: 10.1002/ijc.28948] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/28/2014] [Indexed: 12/11/2022]
Abstract
Loss of BRCA2 function stimulates prostate cancer (PCa) cell invasion and is associated with more aggressive and metastatic tumors in PCa patients. Concurrently, the receptor tyrosine kinase c-kit is highly expressed in skeletal metastases of PCa patients and induced in PCa cells placed into the bone microenvironment in experimental models. However, the precise requirement of c-kit for intraosseous growth of PCa and its relation to BRCA2 expression remain unexplored. Here, we show that c-kit expression promotes migration and invasion of PCa cells. Alongside, we found that c-kit expression in PCa cells parallels BRCA2 downregulation. Gene rescue experiments with human BRCA2 transgene in c-kit-transfected PCa cells resulted in reduction of c-kit protein expression and migration and invasion, suggesting a functional significance of BRCA2 downregulation by c-kit. The inverse association between c-kit and BRCA2 gene expressions in PCa cells was confirmed using laser capture microdissection in experimental intraosseous tumors and bone metastases of PCa patients. Inhibition of bone-induced c-kit expression in PCa cells transduced with lentiviral short hairpin RNA reduced intraosseous tumor incidence and growth. Overall, our results provide evidence of a novel pathway that links bone-induced c-kit expression in PCa cells to BRCA2 downregulation and supports bone metastasis.
Collapse
Affiliation(s)
- Leandro E Mainetti
- Department of Urology, Wayne State University School of Medicine, Detroit, MI
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Elmore MRP, Najafi AR, Koike MA, Dagher NN, Spangenberg EE, Rice RA, Kitazawa M, Matusow B, Nguyen H, West BL, Green KN. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 2014; 82:380-97. [PMID: 24742461 PMCID: PMC4161285 DOI: 10.1016/j.neuron.2014.02.040] [Citation(s) in RCA: 1383] [Impact Index Per Article: 125.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2014] [Indexed: 12/19/2022]
Abstract
The colony-stimulating factor 1 receptor (CSF1R) is a key regulator of myeloid lineage cells. Genetic loss of the CSF1R blocks the normal population of resident microglia in the brain that originates from the yolk sac during early development. However, the role of CSF1R signaling in microglial homeostasis in the adult brain is largely unknown. To this end, we tested the effects of selective CSF1R inhibitors on microglia in adult mice. Surprisingly, extensive treatment results in elimination of ∼99% of all microglia brain-wide, showing that microglia in the adult brain are physiologically dependent upon CSF1R signaling. Mice depleted of microglia show no behavioral or cognitive abnormalities, revealing that microglia are not necessary for these tasks. Finally, we discovered that the microglia-depleted brain completely repopulates with new microglia within 1 week of inhibitor cessation. Microglial repopulation throughout the CNS occurs through proliferation of nestin-positive cells that then differentiate into microglia.
Collapse
Affiliation(s)
- Monica R P Elmore
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - Allison R Najafi
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - Maya A Koike
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - Nabil N Dagher
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - Elizabeth E Spangenberg
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - Rachel A Rice
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA
| | - Masashi Kitazawa
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA 95343, USA
| | | | | | | | - Kim N Green
- Department of Neurobiology and Behavior, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697-4545, USA.
| |
Collapse
|
37
|
Jeffery JJ, Lux K, Vogel JS, Herrera WD, Greco S, Woo HH, AbuShahin N, Pagel MD, Chambers SK. Autocrine inhibition of the c-fms proto-oncogene reduces breast cancer bone metastasis assessed with in vivo dual-modality imaging. Exp Biol Med (Maywood) 2014; 239:404-13. [PMID: 24599884 DOI: 10.1177/1535370214522588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Breast cancer cells preferentially home to the bone microenvironment, which provides a unique niche with a network of multiple bidirectional communications between host and tumor, promoting survival and growth of bone metastases. In the bone microenvironment, the c-fms proto-oncogene that encodes for the CSF-1 receptor, along with CSF-1, serves as one critical cytokine/receptor pair, functioning in paracrine and autocrine fashion. Previous studies concentrated on the effect of inhibition of host (mouse) c-fms on bone metastasis, with resulting decrease in osteolysis and bone metastases as a paracrine effect. In this report, we assessed the role of c-fms inhibition within the tumor cells (autocrine effect) in the early establishment of breast cancer cells in bone and the effects of this early c-fms inhibition on subsequent bone metastases and destruction. This study exploited a multidisciplinary approach by employing two non-invasive, in vivo imaging methods to assess the progression of bone metastases and bone destruction, in addition to ex vivo analyses using RT-PCR and histopathology. Using a mouse model of bone homing human breast cancer cells, we showed that an early one-time application of anti-human c-fms antibody delayed growth of bone metastases and bone destruction for at least 31 days as quantitatively measured by bioluminescence imaging and computed tomography, compared to controls. Thus, neutralizing human c-fms in the breast cancer cell alone decreases extent of subsequent bone metastasis formation and osteolysis. Furthermore, we are the first to show that anti-c-fms antibodies can impact early establishment of breast cancer cells in bone.
Collapse
Affiliation(s)
- Justin J Jeffery
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Esposito M, Kang Y. Targeting tumor-stromal interactions in bone metastasis. Pharmacol Ther 2013; 141:222-33. [PMID: 24140083 DOI: 10.1016/j.pharmthera.2013.10.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 09/05/2013] [Indexed: 12/17/2022]
Abstract
Bone metastasis is a frequent occurrence in late stage solid tumors, including breast cancers, prostate or lung. However, the causes for this proclivity have only recently been elucidated. Significant progress has been made in the past decade toward understanding the molecular underpinnings of bone metastasis, and much of this research reveals a crucial role of the host stroma in each step of the metastatic cascade. Tumor-stromal interactions are crucial in engineering a pre-metastatic niche, accommodating metastatic seeding, and establishing the vicious cycle of bone metastasis. Current treatments in bone metastasis focus on latter steps of the metastatic cascade, with most treatments targeting the process of bone remodeling; however, emerging research identifies many other candidates as promising targets. Host stromal cells including platelets and endothelial cells are important in the early steps of metastatic homing, attachment and extravasation while a variety of immune cells, parenchymal cells and mesenchymal cells of the bone marrow are important in the establishment of overt, immune-suppressed metastatic lesions. Many participants during these steps have been identified and functionally validated. Significant contributors include integrins, (αvβ3, α2β1, α4β1), TGFβ family members, bone resident proteins (BSP, OPG, SPARC, OPN), RANKL, and PTHrP. In this review, we will discuss the contribution of host stromal cells to pre-metastatic niche conditioning, seeding, dormancy, bone-remodeling, immune regulation, and chemotherapeutic shielding in bone metastasis. Research exploring these interactions between bone metastases and stromal cells has yielded many therapeutic targets, and we will discuss both the current and future therapeutic avenues in treating bone metastasis.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, United States.
| |
Collapse
|
39
|
Fend L, Accart N, Kintz J, Cochin S, Reymann C, Le Pogam F, Marchand JB, Menguy T, Slos P, Rooke R, Fournel S, Bonnefoy JY, Préville X, Haegel H. Therapeutic effects of anti-CD115 monoclonal antibody in mouse cancer models through dual inhibition of tumor-associated macrophages and osteoclasts. PLoS One 2013; 8:e73310. [PMID: 24019914 PMCID: PMC3760897 DOI: 10.1371/journal.pone.0073310] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 07/18/2013] [Indexed: 11/18/2022] Open
Abstract
Tumor progression is promoted by Tumor-Associated Macrophages (TAMs) and metastasis-induced bone destruction by osteoclasts. Both myeloid cell types depend on the CD115-CSF-1 pathway for their differentiation and function. We used 3 different mouse cancer models to study the effects of targeting cancer host myeloid cells with a monoclonal antibody (mAb) capable of blocking CSF-1 binding to murine CD115. In mice bearing sub-cutaneous EL4 tumors, which are CD115-negative, the anti-CD115 mAb depleted F4/80+ CD163+ M2-type TAMs and reduced tumor growth, resulting in prolonged survival. In the MMTV-PyMT mouse model, the spontaneous appearance of palpable mammary tumors was delayed when the anti-CD115 mAb was administered before malignant transition and tumors became palpable only after termination of the immunotherapy. When administered to mice already bearing established PyMT tumors, anti-CD115 treatment prolonged their survival and potentiated the effect of chemotherapy with Paclitaxel. As shown by immunohistochemistry, this therapeutic effect correlated with the depletion of F4/80+CD163+ M2-polarized TAMs. In a breast cancer model of bone metastasis, the anti-CD115 mAb potently blocked the differentiation of osteoclasts and their bone destruction activity. This resulted in the inhibition of cancer-induced weight loss. CD115 thus represents a promising target for cancer immunotherapy, since a specific blocking antibody may not only inhibit the growth of a primary tumor through TAM depletion, but also metastasis-induced bone destruction through osteoclast inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sylvie Fournel
- UMR 7199 CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch-Graffenstaden, France
| | | | | | | |
Collapse
|
40
|
Fang H, Declerck YA. Targeting the tumor microenvironment: from understanding pathways to effective clinical trials. Cancer Res 2013; 73:4965-77. [PMID: 23913938 DOI: 10.1158/0008-5472.can-13-0661] [Citation(s) in RCA: 205] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It is clear that tumor cells do not act alone but in close interaction with the extracellular matrix and with stromal cells in the tumor microenvironment (TME). As our understanding of tumor cell-stroma interactions increased over the last two decades, significant efforts have been made to develop agents that interfere with these interactions. Here, we discuss four different therapeutic strategies that target the TME, focusing on agents that are at the most advanced stage of preclinical or clinical development. We end this review by outlining some of the lessons we have learned so far from the development of TME-targeting agents.
Collapse
Affiliation(s)
- Hua Fang
- Division of Hematology-Oncology, University of Southern California, Los Angeles, Los Angeles, USA
| | | |
Collapse
|
41
|
Jia X, Yang FF, Li J, Liu JY, Xue JP. Synthesis and in Vitro Photodynamic Activity of Oligomeric Ethylene Glycol–Quinoline Substituted Zinc(II) Phthalocyanine Derivatives. J Med Chem 2013; 56:5797-805. [DOI: 10.1021/jm400722d] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Xiao Jia
- Fujian Engineering Research Center for Drug and Diagnoses,
Treat of Photodynamic Therapy, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou, China
| | - Feng-Feng Yang
- Fujian Engineering Research Center for Drug and Diagnoses,
Treat of Photodynamic Therapy, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou, China
| | - Jun Li
- Fujian Engineering Research Center for Drug and Diagnoses,
Treat of Photodynamic Therapy, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou, China
| | - Jian-Yong Liu
- Fujian Engineering Research Center for Drug and Diagnoses,
Treat of Photodynamic Therapy, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou, China
| | - Jin-Ping Xue
- Fujian Engineering Research Center for Drug and Diagnoses,
Treat of Photodynamic Therapy, College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou, China
| |
Collapse
|
42
|
Treatment options in castration-resistant prostate cancer: current therapies and emerging docetaxel-based regimens. Urol Oncol 2013; 32:70-9. [PMID: 23506965 DOI: 10.1016/j.urolonc.2013.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 01/28/2023]
Abstract
BACKGROUND Docetaxel-based chemotherapy remains the standard of care for metastatic castration-resistant prostate cancer (mCRPC) and it is the only globally approved first-line therapy. Although docetaxel offers improved survival for this patient population, it is also associated with toxicity and resistance in many patients, representing a need for more efficacious therapies. Preclinical advances have led to improved understanding of the molecular biology of prostate cancer, and targeted therapies that exploit the signaling pathways and molecular targets that underscore the disease are being clinically investigated in combination with docetaxel. DESIGN This article briefly highlights recent data from phase III trials in mCRPC that have led to agent approval. This article also reviews phase II and III trials in which docetaxel-based regimens have been investigated in mCRPC. RESULTS Recently approved agents, including sipuleucel-T, cabazitaxel, abiraterone acetate, and enzalutamide, have diversified the mCRPC treatment landscape. Phase III trials evaluating docetaxel in combination with targeted therapies, including potent oral tyrosine kinase inhibitor, dasatinib, in the READY trial and clusterin inhibitor, custirsen, in the SYNERGY trial, are currently ongoing. CONCLUSIONS In combination with docetaxel, targeted agents dasatinib and custirsen will likely expand the existing treatment paradigm for mCRPC if results from phase III trials are positive.
Collapse
|
43
|
Design and pharmacology of a highly specific dual FMS and KIT kinase inhibitor. Proc Natl Acad Sci U S A 2013; 110:5689-94. [PMID: 23493555 DOI: 10.1073/pnas.1219457110] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Inflammation and cancer, two therapeutic areas historically addressed by separate drug discovery efforts, are now coupled in treatment approaches by a growing understanding of the dynamic molecular dialogues between immune and cancer cells. Agents that target specific compartments of the immune system, therefore, not only bring new disease modifying modalities to inflammatory diseases, but also offer a new avenue to cancer therapy by disrupting immune components of the microenvironment that foster tumor growth, progression, immune evasion, and treatment resistance. McDonough feline sarcoma viral (v-fms) oncogene homolog (FMS) and v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) are two hematopoietic cell surface receptors that regulate the development and function of macrophages and mast cells, respectively. We disclose a highly specific dual FMS and KIT kinase inhibitor developed from a multifaceted chemical scaffold. As expected, this inhibitor blocks the activation of macrophages, osteoclasts, and mast cells controlled by these two receptors. More importantly, the dual FMS and KIT inhibition profile has translated into a combination of benefits in preclinical disease models of inflammation and cancer.
Collapse
|
44
|
Kumari A, Kaur M, Bahia MS, Silakari O. 3D-QSAR analysis of anilinoquinoline inhibitors of colony stimulating factor-1 kinase (cFMS): implementation of field-based molecular alignment. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0513-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
45
|
Francis CL, Kenny PW, Dolezal O, Saubern S, Kruger M, Savage GP, Peat TS, Ryan JH. Construction of the CSIRO Fragment Library. Aust J Chem 2013. [DOI: 10.1071/ch13325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A fundamental component of a successful fragment screening program is a productive fragment library, one that delivers hit fragments with potential for pharmaceutical development. A proprietary fragment library was developed by identifying and extracting subsets of CSIRO’s Compound Collection using two complimentary approaches. Over time, the use of surface plasmon resonance as a front-line screening tool has enabled identification and exclusion of problematic compounds and led to a more reliable fragment screening library.
Collapse
|
46
|
West SD, Goldberg D, Ziegler A, Krencicki M, Du Clos TW, Mold C. Transforming growth factor-β, macrophage colony-stimulating factor and C-reactive protein levels correlate with CD14(high)CD16+ monocyte induction and activation in trauma patients. PLoS One 2012; 7:e52406. [PMID: 23285029 PMCID: PMC3532215 DOI: 10.1371/journal.pone.0052406] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 11/15/2012] [Indexed: 11/19/2022] Open
Abstract
Severe injury remains a leading cause of death and morbidity in patients under 40, with the number of annual trauma-related deaths approaching 160,000 in the United States. Patients who survive the initial trauma and post-traumatic resuscitation are at risk for immune dysregulation, which contributes to late mortality and accounts for approximately 20% of deaths after traumatic injury. This post-traumatic immunosuppressed state has been attributed to over-expression of anti-inflammatory mediators in an effort to restore host homeostasis. We measured a panel of monocyte markers and cytokines in 50 severely injured trauma patients for 3 days following admission. We made the novel observation that the subpopulation of monocytes expressing high levels of CD14 and CD16 was expanded in the majority of patients. These cells also expressed CD163 consistent with differentiation into alternatively activated macrophages with potential regulatory or wound-healing activity. We examined factors in trauma plasma that may contribute to the generation and activation of these cells. The percentage of CD14(high)CD16(+) monocytes after trauma correlated strongly with plasma C-reactive protein (CRP) transforming growth factor-β (TGF-β), and macrophage colony-stimulating factor (M-CSF) levels. We demonstrate a role for TGF-β and M-CSF, but not CRP in generating these cells using monocytes from healthy volunteers incubated with plasma from trauma patients. CD16 is a receptor for CRP and IgG, and we showed that monocytes differentiated to the CD14(high)CD16(+) phenotype produced anti-inflammatory cytokines in response to acute phase concentrations of CRP. The role of these cells in immunosuppression following trauma is an area of ongoing investigation.
Collapse
Affiliation(s)
- Sonlee D West
- Department of Surgery, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.
| | | | | | | | | | | |
Collapse
|
47
|
El-Gamal MI, Anbar HS, Yoo KH, Oh CH. FMS Kinase Inhibitors: Current Status and Future Prospects. Med Res Rev 2012; 33:599-636. [PMID: 22434539 DOI: 10.1002/med.21258] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
FMS, first discovered as the oncogene responsible for Feline McDonough Sarcoma, is a type III receptor tyrosine kinase that binds to the macrophage or monocyte colony-stimulating factor (M-CSF or CSF-1). Signal transduction through that binding results in survival, proliferation, and differentiation of monocyte/macrophage lineage. Overexpression of CSF-1 and/or FMS has been implicated in a number of disease states such as the growth of metastasis of certain types of cancer, in promoting osteoclast proliferation in bone osteolysis, and many inflammatory disorders. Inhibition of CSF-1 and/or FMS may help treat these pathological conditions. This article reviews FMS gene, FMS kinase, CSF-1, IL-34, and their roles in bone osteolysis, cancer biology, and inflammation. Monoclonal antibodies, FMS crystal structure, and small molecule FMS kinase inhibitors of different chemical scaffolds are also reviewed.
Collapse
Affiliation(s)
- Mohammed I El-Gamal
- Biomedical Research Institute, Korea Institute of Science and Technology, P.O. Box 131, Cheongryang, Seoul 130-650, Republic of Korea
| | | | | | | |
Collapse
|
48
|
Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 2012; 122:787-95. [PMID: 22378047 DOI: 10.1172/jci59643] [Citation(s) in RCA: 4638] [Impact Index Per Article: 356.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Diversity and plasticity are hallmarks of cells of the monocyte-macrophage lineage. In response to IFNs, Toll-like receptor engagement, or IL-4/IL-13 signaling, macrophages undergo M1 (classical) or M2 (alternative) activation, which represent extremes of a continuum in a universe of activation states. Progress has now been made in defining the signaling pathways, transcriptional networks, and epigenetic mechanisms underlying M1-M2 or M2-like polarized activation. Functional skewing of mononuclear phagocytes occurs in vivo under physiological conditions (e.g., ontogenesis and pregnancy) and in pathology (allergic and chronic inflammation, tissue repair, infection, and cancer). However, in selected preclinical and clinical conditions, coexistence of cells in different activation states and unique or mixed phenotypes have been observed, a reflection of dynamic changes and complex tissue-derived signals. The identification of mechanisms and molecules associated with macrophage plasticity and polarized activation provides a basis for macrophage-centered diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Antonio Sica
- Istituto Clinico Humanitas IRCCS, Rozzano, Italy.
| | | |
Collapse
|
49
|
Zhong J, Kim MS, Chaerkady R, Wu X, Huang TC, Getnet D, Mitchell CJ, Palapetta SM, Sharma J, O'Meally RN, Cole RN, Yoda A, Moritz A, Loriaux MM, Rush J, Weinstock DM, Tyner JW, Pandey A. TSLP signaling network revealed by SILAC-based phosphoproteomics. Mol Cell Proteomics 2012; 11:M112.017764. [PMID: 22345495 DOI: 10.1074/mcp.m112.017764] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a cytokine that plays diverse roles in the regulation of immune responses. TSLP requires a heterodimeric receptor complex consisting of IL-7 receptor α subunit and its unique TSLP receptor (gene symbol CRLF2) to transmit signals in cells. Abnormal TSLP signaling (e.g. overexpression of TSLP or its unique receptor TSLPR) contributes to the development of a number of diseases including asthma and leukemia. However, a detailed understanding of the signaling pathways activated by TSLP remains elusive. In this study, we performed a global quantitative phosphoproteomic analysis of the TSLP signaling network using stable isotope labeling by amino acids in cell culture. By employing titanium dioxide in addition to antiphosphotyrosine antibodies as enrichment methods, we identified 4164 phosphopeptides on 1670 phosphoproteins. Using stable isotope labeling by amino acids in cell culture-based quantitation, we determined that the phosphorylation status of 226 proteins was modulated by TSLP stimulation. Our analysis identified activation of several members of the Src and Tec families of kinases including Btk, Lyn, and Tec by TSLP for the first time. In addition, we report TSLP-induced phosphorylation of protein phosphatases such as Ptpn6 (SHP-1) and Ptpn11 (Shp2), which has also not been reported previously. Co-immunoprecipitation assays showed that Shp2 binds to the adapter protein Gab2 in a TSLP-dependent manner. This is the first demonstration of an inducible protein complex in TSLP signaling. A kinase inhibitor screen revealed that pharmacological inhibition of PI-3 kinase, Jak family kinases, Src family kinases or Btk suppressed TSLP-dependent cellular proliferation making them candidate therapeutic targets in diseases resulting from aberrant TSLP signaling. Our study is the first phosphoproteomic analysis of the TSLP signaling pathway that greatly expands our understanding of TSLP signaling and provides novel therapeutic targets for TSLP/TSLPR-associated diseases in humans.
Collapse
Affiliation(s)
- Jun Zhong
- McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, 21205 Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Groh J, Weis J, Zieger H, Stanley ER, Heuer H, Martini R. Colony-stimulating factor-1 mediates macrophage-related neural damage in a model for Charcot-Marie-Tooth disease type 1X. Brain 2012; 135:88-104. [PMID: 22094537 PMCID: PMC3267979 DOI: 10.1093/brain/awr283] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/19/2011] [Accepted: 08/26/2011] [Indexed: 12/22/2022] Open
Abstract
Previous studies in our laboratory have shown that in models for three distinct forms of the inherited and incurable nerve disorder, Charcot-Marie-Tooth neuropathy, low-grade inflammation implicating phagocytosing macrophages mediates demyelination and perturbation of axons. In the present study, we focus on colony-stimulating factor-1, a cytokine implicated in macrophage differentiation, activation and proliferation and fostering neural damage in a model for Charcot-Marie-Tooth neuropathy 1B. By crossbreeding a model for the X-linked form of Charcot-Marie-Tooth neuropathy with osteopetrotic mice, a spontaneous null mutant for colony-stimulating factor-1, we demonstrate a robust and persistent amelioration of demyelination and axon perturbation. Furthermore, functionally important domains of the peripheral nervous system, such as juxtaparanodes and presynaptic terminals, were preserved in the absence of colony-stimulating factor-1-dependent macrophage activation. As opposed to other Schwann cell-derived cytokines, colony-stimulating factor-1 is expressed by endoneurial fibroblasts, as revealed by in situ hybridization, immunocytochemistry and detection of β-galactosidase expression driven by the colony-stimulating factor-1 promoter. By both light and electron microscopic studies, we detected extended cell-cell contacts between the colony-stimulating factor-1-expressing fibroblasts and endoneurial macrophages as a putative prerequisite for the effective and constant activation of macrophages by fibroblasts in the chronically diseased nerve. Interestingly, in human biopsies from patients with Charcot-Marie-Tooth type 1, we also found frequent cell-cell contacts between macrophages and endoneurial fibroblasts and identified the latter as main source for colony-stimulating factor-1. Therefore, our study provides strong evidence for a similarly pathogenic role of colony-stimulating factor-1 in genetically mediated demyelination in mice and Charcot-Marie-Tooth type 1 disease in humans. Thus, colony-stimulating factor-1 or its cognate receptor are promising target molecules for treating the detrimental, low-grade inflammation of several inherited neuropathies in humans.
Collapse
Affiliation(s)
- Janos Groh
- Department of Neurology, Section of Developmental Neurobiology, University of Würzburg, Josef-Schneiderstr. 11, 97080 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|