1
|
Tomassetti C, Insinga G, Gimigliano F, Morrione A, Giordano A, Giurisato E. Insights into CSF-1R Expression in the Tumor Microenvironment. Biomedicines 2024; 12:2381. [PMID: 39457693 PMCID: PMC11504891 DOI: 10.3390/biomedicines12102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/30/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The colony-stimulating factor 1 receptor (CSF-1R) plays a pivotal role in orchestrating cellular interactions within the tumor microenvironment (TME). Although the CSF-1R has been extensively studied in myeloid cells, the expression of this receptor and its emerging role in other cell types in the TME need to be further analyzed. This review explores the multifaceted functions of the CSF-1R across various TME cellular populations, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), cancer-associated fibroblasts (CAFs), endothelial cells (ECs), and cancer stem cells (CSCs). The activation of the CSF-1R by its ligands, colony-stimulating factor 1 (CSF-1) and Interleukin-34 (IL-34), regulates TAM polarization towards an immunosuppressive M2 phenotype, promoting tumor progression and immune evasion. Similarly, CSF-1R signaling influences MDSCs to exert immunosuppressive functions, hindering anti-tumor immunity. In DCs, the CSF-1R alters antigen-presenting capabilities, compromising immune surveillance against cancer cells. CSF-1R expression in CAFs and ECs regulates immune modulation, angiogenesis, and immune cell trafficking within the TME, fostering a pro-tumorigenic milieu. Notably, the CSF-1R in CSCs contributes to tumor aggressiveness and therapeutic resistance through interactions with TAMs and the modulation of stemness features. Understanding the diverse roles of the CSF-1R in the TME underscores its potential as a therapeutic target for cancer treatment, aiming at disrupting pro-tumorigenic cellular crosstalk and enhancing anti-tumor immune responses.
Collapse
Affiliation(s)
- Caterina Tomassetti
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Gaia Insinga
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.I.); (F.G.)
| | - Francesca Gimigliano
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy; (G.I.); (F.G.)
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA;
| | - Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
2
|
Moon HG, Eccles JD, Kim SJ, Kim KH, Kim YM, Rehman J, Lee H, Kanabar P, Christman JW, Ackerman SJ, Ascoli C, Kang H, Choi HS, Kim M, You S, Park GY. Complement C1q essential for aeroallergen sensitization via CSF1R + conventional dendritic cells type 2. J Allergy Clin Immunol 2023; 152:1141-1152.e2. [PMID: 37562753 PMCID: PMC10923196 DOI: 10.1016/j.jaci.2023.07.016] [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: 01/27/2023] [Revised: 06/29/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Dendritic cells (DCs) are heterogeneous, comprising multiple subsets with unique functional specifications. Our previous work has demonstrated that the specific conventional type 2 DC subset, CSF1R+cDC2s, plays a critical role in sensing aeroallergens. OBJECTIVE It remains to be understood how CSF1R+cDC2s recognize inhaled allergens. We sought to elucidate the transcriptomic programs and receptor-ligand interactions essential for function of this subset in allergen sensitization. METHODS We applied single-cell RNA sequencing to mouse lung DCs. Conventional DC-selective knockout mouse models were employed, and mice were subjected to inhaled allergen sensitization with multiple readouts of asthma pathology. Under the clinical arm of this work, human lung transcriptomic data were integrated with mouse data, and bronchoalveolar lavage (BAL) specimens were collected from subjects undergoing allergen provocation, with samples assayed for C1q. RESULTS We found that C1q is selectively enriched in lung CSF1R+cDC2s, but not in other lung cDC2 or cDC1 subsets. Depletion of C1q in conventional DCs significantly attenuates allergen sensing and features of asthma. Additionally, we found that C1q binds directly to human dust mite allergen, and the C1q receptor CD91 (LRP1) is required for lung CSF1R+cDC2s to recognize the C1q-allergen complex and induce allergic lung inflammation. Lastly, C1q is enriched in human BAL samples following subsegmental allergen challenge, and human RNA sequencing data demonstrate close homology between lung IGSF21+DCs and mouse CSF1R+cDC2s. CONCLUSIONS C1q is secreted from the CSF1R+cDC2 subset among conventional DCs. Our data indicate that the C1q-LRP1 axis represents a candidate for translational therapeutics in the prevention and suppression of allergic lung inflammation.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago.
| | - Jacob D Eccles
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Seung-Jae Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Ki-Hyun Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Young-Mee Kim
- Department of Pharmacology, University of Illinois College of Medicine, Chicago
| | - Jalees Rehman
- Department of Pharmacology, University of Illinois College of Medicine, Chicago
| | - Hyun Lee
- College of Pharmacy, University of Illinois at Chicago, Chicago
| | - Pinal Kanabar
- Research Informatics Core, University of Illinois at Chicago, Chicago
| | - John W Christman
- Section of Pulmonary, Critical Care, and Sleep Medicine, Columbus; Davis Heart and Lung Research Center, The Ohio State University, Columbus
| | - Steven J Ackerman
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago; Department of Medicine, University of Illinois at Chicago, Chicago
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Minhyung Kim
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles
| | - Sungyong You
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago; Jesse Brown Veterans Affairs Medical Center, Chicago.
| |
Collapse
|
3
|
Kitaba NT, Knudsen GTM, Johannessen A, Rezwan FI, Malinovschi A, Oudin A, Benediktsdottir B, Martino D, González FJC, Gómez LP, Holm M, Jõgi NO, Dharmage SC, Skulstad SM, Watkins SH, Suderman M, Gómez-Real F, Schlünssen V, Svanes C, Holloway JW. Fathers' preconception smoking and offspring DNA methylation. Clin Epigenetics 2023; 15:131. [PMID: 37649101 PMCID: PMC10469907 DOI: 10.1186/s13148-023-01540-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Experimental studies suggest that exposures may impact respiratory health across generations via epigenetic changes transmitted specifically through male germ cells. Studies in humans are, however, limited. We aim to identify epigenetic marks in offspring associated with father's preconception smoking. METHODS We conducted epigenome-wide association studies (EWAS) in the RHINESSA cohort (7-50 years) on father's any preconception smoking (n = 875 offspring) and father's pubertal onset smoking < 15 years (n = 304), using Infinium MethylationEPIC Beadchip arrays, adjusting for offspring age, own smoking and maternal smoking. EWAS of maternal and offspring personal smoking were performed for comparison. Father's smoking-associated dmCpGs were checked in subpopulations of offspring who reported no personal smoking and no maternal smoking exposure. RESULTS Father's smoking commencing preconception was associated with methylation of blood DNA in offspring at two cytosine-phosphate-guanine sites (CpGs) (false discovery rate (FDR) < 0.05) in PRR5 and CENPP. Father's pubertal onset smoking was associated with 19 CpGs (FDR < 0.05) mapped to 14 genes (TLR9, DNTT, FAM53B, NCAPG2, PSTPIP2, MBIP, C2orf39, NTRK2, DNAJC14, CDO1, PRAP1, TPCN1, IRS1 and CSF1R). These differentially methylated sites were hypermethylated and associated with promoter regions capable of gene silencing. Some of these sites were associated with offspring outcomes in this cohort including ever-asthma (NTRK2), ever-wheezing (DNAJC14, TPCN1), weight (FAM53B, NTRK2) and BMI (FAM53B, NTRK2) (p < 0.05). Pathway analysis showed enrichment for gene ontology pathways including regulation of gene expression, inflammation and innate immune responses. Father's smoking-associated sites did not overlap with dmCpGs identified in EWAS of personal and maternal smoking (FDR < 0.05), and all sites remained significant (p < 0.05) in analyses of offspring with no personal smoking and no maternal smoking exposure. CONCLUSION Father's preconception smoking, particularly in puberty, is associated with offspring DNA methylation, providing evidence that epigenetic mechanisms may underlie epidemiological observations that pubertal paternal smoking increases risk of offspring asthma, low lung function and obesity.
Collapse
Affiliation(s)
- Negusse Tadesse Kitaba
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Gerd Toril Mørkve Knudsen
- Department of Clinical Sciences, University of Bergen, Bergen, Norway
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - Ane Johannessen
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Faisal I Rezwan
- Department of Computer Science, Aberystwyth University, Aberystwyth, UK
| | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Anna Oudin
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bryndis Benediktsdottir
- Department of Allergy, Respiratory Medicine and Sleep, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - David Martino
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | | | | | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Nils Oskar Jõgi
- Department of Clinical Sciences, University of Bergen, Bergen, Norway
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Svein Magne Skulstad
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - Sarah H Watkins
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Matthew Suderman
- University of Bristol, MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, Bristol, UK
| | - Francisco Gómez-Real
- Department of Clinical Sciences, University of Bergen, Bergen, Norway
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Vivi Schlünssen
- Department of Public Health, Work, Environment and Health, Danish Ramazzini Centre, Aarhus University Denmark, Aarhus, Denmark
- National Research Center for the Working Environment, Copenhagen, Denmark
| | - Cecilie Svanes
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.
- NIHR Southampton Biomedical Research Center, University Hospitals Southampton, Southampton, UK.
| |
Collapse
|
4
|
Du L, Tang L, Xiao L, Tang K, Zeng Z, Liang Y, Guo Y. Increased expression of CSF1 in patients with eosinophilic asthma. Immun Inflamm Dis 2023; 11:e847. [PMID: 37249291 PMCID: PMC10170305 DOI: 10.1002/iid3.847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/29/2023] [Accepted: 04/10/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The link between colony-stimulating factor 1 (CSF1) and asthma was reported recently. However, the role and mechanism of CSF1 in asthma remain poorly understood. In this study, we aimed to explore the expression and its potential mechanism of CSF1 in asthma. METHODS CSF1 expression in the airway samples from asthmatics and healthy controls were examined, then the correlations between CSF1 and eosinophilic indicators were analyzed. Subsequently, bronchial epithelial cells (BEAS-2B) with CSF1 overexpression and knockdown were constructed to investigate the potential molecular mechanism of CSF1. Finally, the effect of CSF1R inhibitor on STAT1 was investigated. RESULTS The expression of CSF1 was significantly increased in patients with asthma compared to healthy controls, especially in patients with severe and eosinophilic asthma. Upregulated CSF1 positively correlated with airway-increased eosinophil inflammation. In vitro, cytokines interleukin 13 (IL-13) and IL-33 can stimulate the upregulation of CSF1 expression. CSF1 overexpression enhanced p-CSF1R/CSF1R and p-STAT1/STAT1 expression, while knockdown CSF1 using anti-CSF1 siRNAs decreased p-CSF1R/CSF1R and p-STAT1/STAT1 expression. Furthermore, the inhibitor of CSF1R significantly decreased p-STAT1/STAT1 expression. CONCLUSIONS Sputum CSF1 may be involved in asthmatic airway eosinophil inflammation by interacting with CSF1R and further activating the STAT1 signaling. Interfering this potential pathway could serve as an anti-inflammatory therapy for asthma.
Collapse
Affiliation(s)
- Lijuan Du
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Lu Tang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Lisha Xiao
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Kun Tang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Zhimin Zeng
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yuxia Liang
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Yubiao Guo
- Department of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
- Institute of Respiratory Diseases of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
5
|
Xiang C, Li H, Tang W. Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res 2023; 187:106566. [PMID: 36423789 DOI: 10.1016/j.phrs.2022.106566] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Colony-stimulating factor-1 receptor (CSF-1R), also known as FMS kinase, is a type I single transmembrane protein mainly expressed in myeloid cells, such as monocytes, macrophages, glial cells, and osteoclasts. The endogenous ligands, colony-stimulating factor-1 (CSF-1) and Interleukin-34 (IL-34), activate CSF-1R and downstream signaling pathways including PI3K-AKT, JAK-STATs, and MAPKs, and modulate the proliferation, differentiation, migration, and activation of target immune cells. Over the past decades, the promising therapeutic potential of CSF-1R signaling inhibition has been widely studied for decreasing immune suppression and escape in tumors, owing to depletion and reprogramming of tumor-associated macrophages. In addition, the excessive activation of CSF-1R in inflammatory diseases is consecutively uncovered in recent years, which may result in inflammation in bone, kidney, lung, liver and central nervous system. Agents against CSF-1R signaling have been increasingly investigated in preclinical or clinical studies for inflammatory diseases treatment. However, the pathological mechanism of CSF-1R in inflammation is indistinct and whether CSF-1R signaling can be identified as biomarkers remains controversial. With the background information aforementioned, this review focus on the dialectical roles of CSF-1R and its ligands in regulating innate immune cells and highlights various therapeutic implications of blocking CSF-1R signaling in inflammatory diseases.
Collapse
Affiliation(s)
- Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Xiang C, Fan C, Lu Q, Liu M, Lu H, Feng C, Wu Y, Wu B, Li H, Tang W. Interfering with alternatively activated macrophages by CSF-1R inhibition exerts therapeutic capacity on allergic airway inflammation. Biochem Pharmacol 2022; 198:114952. [PMID: 35149050 DOI: 10.1016/j.bcp.2022.114952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE Allergic asthma is a chronic inflammatory disorder with airway hyperresponsiveness and tissue remodeling as the main pathological characteristics. The etiology of asthma is relatively complicated, involving genetic susceptibility, epigenetic regulation, environmental factors, and immune imbalance. Colony stimulating factor 1 receptor (CSF-1R), highly expressed in myeloid monocytes, plays an important role in regulating inflammation. However, the pathological role of CSF-1R and the therapeutic effects of CSF-1R inhibitor in allergic airway inflammation remain indistinct. METHODS The house dust mite (HDM)-triggered allergic airway inflammation model was conducted to fully uncover the efficacies of CSF-1R inhibition, as illustrated by histopathological examinations, biochemical analysis, ELISA, RT-PCR, Western blotting assay, immunofluorescence, and flow cytometry. Furthermore, bone marrow-derived macrophages (BMDMs) were differentiated and polarized upon IL-4/IL-13 induction to clarify the underlying mechanisms of CSF-1R inhibition. RESULTS Herein, we presented that the expression of CSF-1R was increased in HDM-induced experimental asthma and inhibition of CSF-1R displayed dramatic effects on the disease severity of asthma, referring to suppressing the secretion of allergic mediators, dysfunction of airway epithelium, and infiltration of inflammatory cells. Furthermore, CSF-1R inhibitor could markedly restrain the polarization and expression of transcriptional factors of alternatively activated macrophages (AAMs) in the presence of IL-4/IL-13 and reduce the recruitment of CSF-1R-dominant macrophages, both in acute and chronic allergic airway inflammation model. CONCLUSION Collectively, our findings demonstrated the molecular pathological mechanism of CSF-1R in allergic airway diseases and suggested that targeting CSF-1R might be an alternative intervention strategy on the homeostasis of airway immune microenvironment in asthma.
Collapse
Affiliation(s)
- Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Fan
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qiukai Lu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moting Liu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huimin Lu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunlan Feng
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yanwei Wu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Wu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
Moon HG, Kim SJ, Lee MK, Kang H, Choi HS, Harijith A, Ren J, Natarajan V, Christman JW, Ackerman SJ, Park GY. Colony-stimulating factor 1 and its receptor are new potential therapeutic targets for allergic asthma. Allergy 2020; 75:357-369. [PMID: 31385613 PMCID: PMC7002247 DOI: 10.1111/all.14011] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/10/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND A new approach targeting aeroallergen sensing in the early events of mucosal immunity could have greater benefit. The CSF1-CSF1R pathway has a critical role in trafficking allergens to regional lymph nodes through activating dendritic cells. Intervention in this pathway could prevent allergen sensitization and subsequent Th2 allergic inflammation. OBJECTIVE To examine the therapeutic effectiveness of CSF1 and CSF1R inhibition for blocking the dendritic cell function of sensing aeroallergens. METHODS We adopted a model of chronic asthma induced by a panel of three naturally occurring allergens and novel delivery system of CSF1R inhibitor encapsulated nanoprobe. RESULTS Selective depletion of CSF1 in airway epithelial cells abolished the production of allergen-reactive IgE, resulting in prevention of new asthma development as well as reversal of established allergic lung inflammation. CDPL-GW nanoprobe containing GW2580, a selective CSF1R inhibitor, showed favorable pharmacokinetics for inhalational treatment and intranasal insufflation delivery of CDPL-GW nanoprobe ameliorated asthma pathologies including allergen-specific serum IgE production, allergic lung and airway inflammation and airway hyper-responsiveness (AHR) with minimal pulmonary adverse reaction. CONCLUSION The inhibition of the CSF1-CSF1R signaling pathway effectively suppresses sensitization to aeroallergens and consequent allergic lung inflammation in a murine model of chronic asthma. CSF1R inhibition is a promising new target for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Seung-jae Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Myoung Kyu Lee
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Homan Kang
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anantha Harijith
- Department of Pediatrics, University of Illinois at Chicago, IL, USA
| | - Jinhong Ren
- Center for Biomolecular Science, College of Pharmacy, University of Illinois at Chicago, IL, USA
| | - Viswanathan Natarajan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Department of Pharmacology, University of Illinois at Chicago, IL, USA
| | - John W. Christman
- Section of Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Center, Columbus, Ohio, USA
| | - Steven J. Ackerman
- Department of Biochemistry and Molecular Genetics, and Medicine, University of Illinois at Chicago, IL, USA
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
8
|
Association between CSF1 and CSF1R Polymorphisms and Parkinson's Disease in Taiwan. J Clin Med 2019; 8:jcm8101529. [PMID: 31554150 PMCID: PMC6832167 DOI: 10.3390/jcm8101529] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/15/2019] [Accepted: 09/16/2019] [Indexed: 12/26/2022] Open
Abstract
Background: CSF1/CSF1R neuroinflammatory signaling is emerging as an important pathway involved in the pathogenesis of Parkinson’s disease (PD). However, the genetic associations between CSF1/CSF1R and PD have not yet been explored. Methods: We investigated the effects of two functional genetic variants, including CSF1 rs1058885 and CSF1R rs10079250 in a cohort including 502 Taiwanese patients with PD and 511 age- and gender-matched healthy controls. Results: The CSF1 rs1058885 TT genotype was less frequent in PD patients compared with control subjects (odds ratio (OR) = 0.63, 95% confidence interval (CI): 0.43–0.92, p = 0.015). The PD patients also had a lower frequency of the CSF1 rs1058885 T allele compared with the control subjects (OR = 0.80, 95% CI: 0.67–0.96, p = 0.014). No statistically significant differences in allelic and genotypic frequencies of CSF1R rs10079250 between the PD and control subjects were found, even after stratification by age at onset and gender. Conclusion: This study reports a genetic association between CSF1 and PD for the first time.
Collapse
|
9
|
Zhao M, Zhang Y, Liu Y, Sun G, Tian H, Hong L. Polymorphisms in MAPK9 (rs4147385) and CSF1R (rs17725712) are associated with the development of inhibitors in patients with haemophilia A in North China. Int J Lab Hematol 2019; 41:572-577. [PMID: 31149782 DOI: 10.1111/ijlh.13055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/28/2019] [Accepted: 05/06/2019] [Indexed: 02/03/2023]
Abstract
INTRODUCTION The formation of neutralizing antibodies (FVIII inhibitors) in haemophilia A patients is an immune response to the deficient factor. This process is multifactorial and includes environmental and genetic factors. Some genetic markers that play a decisive role in the immune response have been identified as risk factors for inhibitor development. OBJECTIVE Our aim was to investigate the association between polymorphisms in several genes involved in the regulation of the immune response and inhibitor development in patients with haemophilia A in North China. METHODS We analysed eight SNPs (MAPK9 rs4147385, CSF1R rs17725712, CD44 rs927335, STAT4 rs7574865, IKZF1 rs4917014, ETS1 rs6590330, BANK1 rs17266594 and rs10516487) by Snapshot SNP genotyping assays in 100 haemophilia A patients, including 29 with inhibitors and 71 without inhibitors. RESULTS Our results demonstrated that the rs17725712 A allele and the AA homozygous genotype of CSF1R were more frequent in patients with inhibitors. The rs4147385 G allele in MAPK9 was also more frequent in the inhibitor cohort. CONCLUSION We confirmed an association of CSF1R rs17725712 and MAPK9 rs4147385 with inhibitor development in haemophilia A patients in North China.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yujing Zhang
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yanyan Liu
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guoxun Sun
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hong Tian
- Clinical Laboratory Department, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Luojia Hong
- Department of Hematology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Moon HG, Kim SJ, Jeong JJ, Han SS, Jarjour NN, Lee H, Abboud-Werner SL, Chung S, Choi HS, Natarajan V, Ackerman SJ, Christman JW, Park GY. Airway Epithelial Cell-Derived Colony Stimulating Factor-1 Promotes Allergen Sensitization. Immunity 2018; 49:275-287.e5. [PMID: 30054206 DOI: 10.1016/j.immuni.2018.06.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/25/2018] [Accepted: 06/21/2018] [Indexed: 10/28/2022]
Abstract
Airway epithelial cells (AECs) secrete innate immune cytokines that regulate adaptive immune effector cells. In allergen-sensitized humans and mice, the airway and alveolar microenvironment is enriched with colony stimulating factor-1 (CSF1) in response to allergen exposure. In this study we found that AEC-derived CSF1 had a critical role in the production of allergen reactive-IgE production. Furthermore, spatiotemporally secreted CSF1 regulated the recruitment of alveolar dendritic cells (DCs) and enhanced the migration of conventional DC2s (cDC2s) to the draining lymph node in an interferon regulatory factor 4 (IRF4)-dependent manner. CSF1 selectively upregulated the expression of the chemokine receptor CCR7 on the CSF1R+ cDC2, but not the cDC1, population in response to allergen stimuli. Our data describe the functional specification of CSF1-dependent DC subsets that link the innate and adaptive immune responses in T helper 2 (Th2) cell-mediated allergic lung inflammation.
Collapse
Affiliation(s)
- Hyung-Geun Moon
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Seung-Jae Kim
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jong Jin Jeong
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Seon-Sook Han
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Nizar N Jarjour
- Allergy, Pulmonary, and Critical Care Division, Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Hyun Lee
- Center for Biomolecular Sciences, and Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Sherry L Abboud-Werner
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sangwoon Chung
- Section of Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Center, Columbus, OH, USA
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Viswanathan Natarajan
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven J Ackerman
- Department of Biochemistry and Molecular Genetics, and Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - John W Christman
- Section of Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Center, Columbus, OH, USA
| | - Gye Young Park
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
11
|
Guillonneau C, Bézie S, Anegon I. Immunoregulatory properties of the cytokine IL-34. Cell Mol Life Sci 2017; 74:2569-2586. [PMID: 28258292 PMCID: PMC11107603 DOI: 10.1007/s00018-017-2482-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/10/2017] [Accepted: 01/30/2017] [Indexed: 12/21/2022]
Abstract
Interleukin-34 is a cytokine with only partially understood functions, described for the first time in 2008. Although IL-34 shares very little homology with CSF-1 (CSF1, M-CSF), they share a common receptor CSF-1R (CSF-1R) and IL-34 has also two distinct receptors (PTP-ζ) and CD138 (syndecan-1). To make the situation more complex, IL-34 has also been shown as pairing with CSF-1 to form a heterodimer. Until now, studies have demonstrated that this cytokine is released by some tissues that differ to those where CSF-1 is expressed and is involved in the differentiation and survival of macrophages, monocytes, and dendritic cells in response to inflammation. The involvement of IL-34 has been shown in areas as diverse as neuronal protection, autoimmune diseases, infection, cancer, and transplantation. Our recent work has demonstrated a new and possible therapeutic role for IL-34 as a Foxp3+ Treg-secreted cytokine mediator of transplant tolerance. In this review, we recapitulate most recent findings on IL-34 and its controversial effects on immune responses and address its immunoregulatory properties and the potential of targeting this cytokine in human.
Collapse
Affiliation(s)
- Carole Guillonneau
- INSERM UMR1064, Center for Research in Transplantation and Immunology-ITUN, Université de Nantes, 30 Bd. Jean Monnet, 44093, Nantes Cedex 01, France.
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.
| | - Séverine Bézie
- INSERM UMR1064, Center for Research in Transplantation and Immunology-ITUN, Université de Nantes, 30 Bd. Jean Monnet, 44093, Nantes Cedex 01, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Ignacio Anegon
- INSERM UMR1064, Center for Research in Transplantation and Immunology-ITUN, Université de Nantes, 30 Bd. Jean Monnet, 44093, Nantes Cedex 01, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
12
|
Pridans C, Davis GM, Sauter KA, Lisowski ZM, Corripio-Miyar Y, Raper A, Lefevre L, Young R, McCulloch ME, Lillico S, Milne E, Whitelaw B, Hume DA. A Csf1r-EGFP Transgene Provides a Novel Marker for Monocyte Subsets in Sheep. THE JOURNAL OF IMMUNOLOGY 2016; 197:2297-305. [PMID: 27521343 PMCID: PMC5009875 DOI: 10.4049/jimmunol.1502336] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/15/2016] [Indexed: 12/12/2022]
Abstract
Expression of Csf1r in adults is restricted to cells of the macrophage lineage. Transgenic reporters based upon the Csf1r locus require inclusion of the highly conserved Fms-intronic regulatory element for expression. We have created Csf1r-EGFP transgenic sheep via lentiviral transgenesis of a construct containing elements of the mouse Fms-intronic regulatory element and Csf1r promoter. Committed bone marrow macrophage precursors and blood monocytes express EGFP in these animals. Sheep monocytes were divided into three populations, similar to classical, intermediate, and nonclassical monocytes in humans, based upon CD14 and CD16 expression. All expressed EGFP, with increased levels in the nonclassical subset. Because Csf1r expression coincides with the earliest commitment to the macrophage lineage, Csf1r-EGFP bone marrow provides a tool for studying the earliest events in myelopoiesis using the sheep as a model.
Collapse
Affiliation(s)
- Clare Pridans
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Gemma M Davis
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Kristin A Sauter
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Zofia M Lisowski
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | | | - Anna Raper
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Lucas Lefevre
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Rachel Young
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Mary E McCulloch
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Simon Lillico
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Elspeth Milne
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Bruce Whitelaw
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - David A Hume
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| |
Collapse
|
13
|
Toh ML, Bonnefoy JY, Accart N, Cochin S, Pohle S, Haegel H, De Meyer M, Zemmour C, Preville X, Guillen C, Thioudellet C, Ancian P, Lux A, Sehnert B, Nimmerjahn F, Voll RE, Schett G. Bone- and Cartilage-Protective Effects of a Monoclonal Antibody Against Colony-Stimulating Factor 1 Receptor in Experimental Arthritis. Arthritis Rheumatol 2014; 66:2989-3000. [DOI: 10.1002/art.38624] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/06/2014] [Indexed: 01/08/2023]
Affiliation(s)
| | | | | | | | - Sandy Pohle
- University of Erlangen-Nuremberg; Erlangen Germany
| | | | | | | | | | | | | | | | - Anja Lux
- University of Erlangen-Nuremberg; Erlangen Germany
| | | | | | | | - Georg Schett
- University of Erlangen-Nuremberg; Erlangen Germany
| |
Collapse
|
14
|
Kim JH, Park BL, Cheong HS, Bae JS, Kim LH, Kim JW, Lee BC, Seo CH, Kang TC, Park SH, Choi IG, Shin HD. Association BetweenHTR7Genetic Polymorphisms and Alcohol Dependence, Using the Alcohol Use Disorders Identification Test (AUDIT). Alcohol Clin Exp Res 2014; 38:2354-61. [DOI: 10.1111/acer.12482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 05/05/2014] [Indexed: 12/16/2022]
Affiliation(s)
- Jeong-Hyun Kim
- Department of Life Science; Sogang University; Seoul Korea
- Research Institute for Basic Science; Sogang University; Seoul Korea
| | - Byung-Lae Park
- Department of Genetic Epidemiology; SNP Genetics, Inc.; Seoul Korea
| | - Hyun Sub Cheong
- Department of Genetic Epidemiology; SNP Genetics, Inc.; Seoul Korea
| | - Joon Seol Bae
- Laboratory of Translational Genomics; Samsung Medical Center; Samsung Genome Institute; Seoul Korea
| | - Lyoung Hyo Kim
- Department of Life Science; Sogang University; Seoul Korea
- Department of Genetic Epidemiology; SNP Genetics, Inc.; Seoul Korea
| | - Jee Wook Kim
- Department of Neuropsychiatry; Hallym University Dongtan Sacred Heart Hospital; Hwaseong Korea
- Burn Institute; Hallym University; Seoul Korea
| | - Byoung Chul Lee
- Burn Institute; Hallym University; Seoul Korea
- Department of Neuropsychiatry; Hangang Sacred Heart Hospital; Hallym University; Seoul Korea
| | - Cheong Hoon Seo
- Burn Institute; Hallym University; Seoul Korea
- Department of Rehabilitation Medicine; Hangang Sacred Heart Hospital; Hallym University; Seoul Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology; Institute of Epilepsy Research; College of Medicine; Hallym University; Chuncheon Korea
| | - Sun-Hee Park
- Keyo Medical Foundation; Keyo Hospital; Uiwang Korea
| | - Ihn-Geun Choi
- Burn Institute; Hallym University; Seoul Korea
- Department of Neuropsychiatry; Hallym University Kangnam Sacred Heart Hospital; Seoul Korea
| | - Hyoung Doo Shin
- Department of Life Science; Sogang University; Seoul Korea
- Research Institute for Basic Science; Sogang University; Seoul Korea
| |
Collapse
|
15
|
Börnigen D, Tranchevent LC, Bonachela-Capdevila F, Devriendt K, De Moor B, De Causmaecker P, Moreau Y. An unbiased evaluation of gene prioritization tools. Bioinformatics 2012; 28:3081-8. [PMID: 23047555 DOI: 10.1093/bioinformatics/bts581] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
MOTIVATION Gene prioritization aims at identifying the most promising candidate genes among a large pool of candidates-so as to maximize the yield and biological relevance of further downstream validation experiments and functional studies. During the past few years, several gene prioritization tools have been defined, and some of them have been implemented and made available through freely available web tools. In this study, we aim at comparing the predictive performance of eight publicly available prioritization tools on novel data. We have performed an analysis in which 42 recently reported disease-gene associations from literature are used to benchmark these tools before the underlying databases are updated. RESULTS Cross-validation on retrospective data provides performance estimate likely to be overoptimistic because some of the data sources are contaminated with knowledge from disease-gene association. Our approach mimics a novel discovery more closely and thus provides more realistic performance estimates. There are, however, marked differences, and tools that rely on more advanced data integration schemes appear more powerful. CONTACT yves.moreau@esat.kuleuven.be SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Daniela Börnigen
- Department of Electrical Engineering, ESAT-SCD, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
16
|
Therapeutic applications of macrophage colony-stimulating factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood 2011; 119:1810-20. [PMID: 22186992 DOI: 10.1182/blood-2011-09-379214] [Citation(s) in RCA: 559] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Macrophage-colony stimulating factor (CSF-1) signaling through its receptor (CSF-1R) promotes the differentiation of myeloid progenitors into heterogeneous populations of monocytes, macrophages, dendritic cells, and bone-resorbing osteoclasts. In the periphery, CSF-1 regulates the migration, proliferation, function, and survival of macrophages, which function at multiple levels within the innate and adaptive immune systems. Macrophage populations elicited by CSF-1 are associated with, and exacerbate, a broad spectrum of pathologies, including cancer, inflammation, and bone disease. Conversely, macrophages can also contribute to immunosuppression, disease resolution, and tissue repair. Recombinant CSF-1, antibodies against the ligand and the receptor, and specific inhibitors of CSF-1R kinase activity have been each been tested in a range of animal models and in some cases, in patients. This review examines the potential clinical uses of modulators of the CSF-1/CSF-1R system. We conclude that CSF-1 promotes a resident-type macrophage phenotype. As a treatment, CSF-1 has therapeutic potential in tissue repair. Conversely, inhibition of CSF-1R is unlikely to be effective in inflammatory disease but may have utility in cancer.
Collapse
|
17
|
Park CS, Rhim T. Application of proteomics in asthma research. Expert Rev Proteomics 2011; 8:221-30. [PMID: 21501015 DOI: 10.1586/epr.11.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bronchial asthma is caused by allergic airway inflammation, resulting in reversible airway obstruction, characterized by airway hyper-responsiveness, bronchoconstriction, increased mucus secretion and an increase in lung vessel permeability. The pathophysiological changes in asthma have been attributed to the altered expression of biologically plausible proteins associated with transcriptional pathways, inflammatory mediators, chemokines, cytokines, apoptosis and cell proliferation. Such multifactorial diseases characteristically involve an interplay of many genetic variations of molecular and biochemical pathways and their interactions with environmental factors. The complex nature of the asthma phenotype, together with genetic heterogeneity and environmental influences, has made it difficult to uncover the aspects that underlie this common disease. Recently, genomic and proteomic technologies have been developed to identify associations between genes, proteins and disease. This approach, called 'omics biology', aims to recognize early onset of disease, institute preventive treatment and identify new molecular targets for novel drugs in multifactorial diseases. This article reviews examples of how proteomic technology can be used to find asthma marker proteins (from the cell model to clinical samples). Identification of protein changes in different stages of asthma could provide further insights into the complex molecular mechanisms involved in this disease. These studies provide new insights for finding novel pathological mediators and biomarkers of asthma.
Collapse
Affiliation(s)
- Choon-Sik Park
- Genome Research Center for Allergy and Respiratory Disease, Soonchunhyang University Hospital, Bucheon, South Korea
| | | |
Collapse
|
18
|
Cervantes S, Samaranch L, Vidal-Taboada JM, Lamet I, Bullido MJ, Frank-García A, Coria F, Lleó A, Clarimón J, Lorenzo E, Alonso E, Sánchez-Juan P, Rodríguez-Rodríguez E, Combarros O, Rosich M, Vilella E, Pastor P. Genetic variation in APOE cluster region and Alzheimer's disease risk. Neurobiol Aging 2011; 32:2107.e7-17. [PMID: 21752496 DOI: 10.1016/j.neurobiolaging.2011.05.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 05/10/2011] [Accepted: 05/30/2011] [Indexed: 11/29/2022]
Abstract
We report the fine mapping/sequencing results of promoter and regulatory regions of APOE cluster genes (APOE, APOC1, APOC4, APOC2, and TOMM40) in Alzheimer's disease (AD) risk as well as in the progression from mild cognitive impairment (MCI) to AD. Long-range sequencing in 29 MCI subjects who progressed to dementia revealed 7 novel variants. Two potentially relevant novel variants and 34 single nucleotide polymorphisms (SNPs) were genotyped in a large sample of AD, MCI, and control subjects (n = 1453). Globally, very little association signal was observed in our sample in the absence of APOE ε4. Rs5158 (APOC4 intron 1) and rs10413089 (3' to APOC2) showed a trend toward an increase in AD risk independently from APOE ε4 associated risk though it did not survive multiple test correction (uncorrected p = 0.0099 and 0.01, respectively). Interestingly, rs10413089 showed a similar effect in an independent series. The analysis of the discovery sample showed an association of TOMM40 single nucleotide polymorphisms with progression from MCI stage to AD (rs59007384 and rs11556510), as well as with a shorter time to progression from MCI status to AD (rs10119), though these results could not be replicated in independent series. Further studies are needed to investigate the role of APOE cluster variants in AD risk.
Collapse
Affiliation(s)
- Sebastián Cervantes
- Neurogenetics Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Lee SH, Park JS, Park CS. The search for genetic variants and epigenetics related to asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2011; 3:236-44. [PMID: 21966603 PMCID: PMC3178821 DOI: 10.4168/aair.2011.3.4.236] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/29/2011] [Indexed: 12/12/2022]
Abstract
For the past two decades, a huge number of genetic studies have been conducted to identify the genetic variants responsible for asthma risk. Several types of genetic and genomic approaches, including linkage analysis, candidate gene single nucleotide polymorphism studies, and whole genome-wide association studies have been applied. In this review article, the results of these approaches are summarized, and their limitations are discussed. Additionally, perspectives for applying upcoming new epigenetic or genomic technologies, such as copy number variation, are introduced to increase our understanding of new omic approaches to asthma genetics.
Collapse
Affiliation(s)
- Shin-Hwa Lee
- Genome Research Center for Allergy and Respiratory Disease, Division of Allergy and Respiratory Medicine, Soonchunhyang University Hospital, Bucheon, Korea
| | | | | |
Collapse
|