1
|
Perçin G, Riege K, Fröbel J, Metz J, Culemann S, Lesche M, Reinhardt S, Höfer T, Hoffmann S, Waskow C. Embryonic macrophages orchestrate niche cell homeostasis for the establishment of the definitive hematopoietic stem cell pool. Nat Commun 2025; 16:4428. [PMID: 40368907 PMCID: PMC12078706 DOI: 10.1038/s41467-025-59059-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 03/25/2025] [Indexed: 05/16/2025] Open
Abstract
Embryonic macrophages emerge before the onset of definitive hematopoiesis, seed into discrete tissues and contribute to specialized resident macrophages throughout life. Presence of embryonic macrophages in the bone marrow and functional impact on hematopoietic stem cells (HSC) or the niche remains unclear. Here we show that bone marrow macrophages consist of two ontogenetically distinct cell populations from embryonic and adult origin. Newborn mice lacking embryonic macrophages have decreased HSC numbers in the bone marrow suggesting an important function for embryo-derived macrophages in orchestrating HSC trafficking around birth. The establishment of a normal cellular niche space in the bone marrow critically depends on embryonic macrophages that are important for the development of mesenchymal stromal cells, but not other non-hematopoietic niche cells, providing evidence for a specific role for embryo-derived macrophages in the establishment of the niche environment pivotal for the establishment of a normally sized HSC pool.
Collapse
Affiliation(s)
- Gülce Perçin
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
| | - Konstantin Riege
- Computational Biology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Julia Fröbel
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Jonas Metz
- Theoretical Systems Biology, German Cancer Research Center, Heidelberg, Germany
| | - Stephan Culemann
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Mathias Lesche
- DRESDEN-concept Genome Center, c/o CMCB Center for Molecular and Cellular Bioengineering Technology Platform of the TUD Dresden University of Technology, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, c/o CMCB Center for Molecular and Cellular Bioengineering Technology Platform of the TUD Dresden University of Technology, Dresden, Germany
| | - Thomas Höfer
- Theoretical Systems Biology, German Cancer Research Center, Heidelberg, Germany
| | - Steve Hoffmann
- Computational Biology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany
| | - Claudia Waskow
- Immunology of Aging, Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University, Jena, Germany.
- Department of Medicine III, Faculty of Medicine, TU Dresden, Dresden, Germany.
| |
Collapse
|
2
|
Weinhaus B, Homan S, Kincaid M, Tadwalkar A, Gu X, Kumar S, Slaughter A, Zhang J, Wu Q, Kofron JM, Troutman TD, DeFalco T, Lucas D. Differential regulation of fetal bone marrow and liver hematopoiesis by yolk-sac-derived myeloid cells. Nat Commun 2025; 16:4427. [PMID: 40368906 PMCID: PMC12078524 DOI: 10.1038/s41467-025-59058-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Fetal hematopoiesis takes place in the liver before colonizing the bone marrow where it will persist for life. This colonization is thought to be mediated by specification of a microenvironment that selectively recruits hematopoietic cells to the nascent bone marrow. The identity and mechanisms regulating the specification of this colonization niche are unclear. Here we identify a VCAM1+ sinusoidal colonization niche in the diaphysis that regulates neutrophil and hematopoietic stem cell colonization of the bone marrow. Using confocal microscopy, we find that colonizing hematopoietic stem and progenitor cells (HSPC) and myeloid cells selectively localize to a subset of VCAM1+ sinusoids in the center of the diaphysis. Vcam1 deletion in endothelial cells impairs hematopoietic colonization while depletion of yolk-sac-derived osteoclasts disrupts VCAM1+ expression, and impairs neutrophil and HSPC colonization to the bone marrow. Depletion of yolk-sac-derived myeloid cells increases fetal liver hematopoietic stem cell numbers, function and erythropoiesis independent of osteoclast activity. Thus, the yolk sac produces myeloid cells that have opposite roles in fetal hematopoiesis: while yolk-sac derived myeloid cells in the bone marrow promote hematopoietic colonization by specifying a VCAM1+ colonization niche, a different subset of yolk-sac-derived myeloid cells inhibits HSC in the fetal liver.
Collapse
Affiliation(s)
- Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Shelli Homan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Morgan Kincaid
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Aryan Tadwalkar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Sumit Kumar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
| | - Anastasiya Slaughter
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Jizhou Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Qingqing Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - J Matthew Kofron
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Ty D Troutman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
3
|
Passin V, Ledesma‐Colunga MG, Altamura S, Muckenthaler MU, Baschant U, Hofbauer LC, Rauner M. Depletion of macrophages and osteoclast precursors mitigates iron overload-mediated bone loss. IUBMB Life 2025; 77:e2928. [PMID: 39555707 PMCID: PMC11611226 DOI: 10.1002/iub.2928] [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: 08/12/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Iron is an essential element for physiological cellular processes, but is toxic in excess. Iron overload diseases are commonly associated with low bone mass. Increased bone resorption by osteoclasts as well as decreased bone formation by osteoblasts have been implicated in bone loss under iron overload conditions. However, the exact contribution of individual cell types has not yet been formally tested. In this study, we aimed to investigate the role of osteoclast precursors in iron overload-induced bone loss. To that end, we used clodronate liposomes to deplete phagocytic cells (including macrophages and osteoclast precursors) in male C57BL/6J mice that were exposed to ferric derisomaltose. Bone microarchitecture and bone turnover were assessed after 4 weeks. The application of clodronate resulted in the efficient depletion of circulating myeloid-lineage cells by about 70%. Depletion of osteoclast precursors mitigated iron overload-induced trabecular bone loss at the lumbar vertebrae and distal femur. While clodronate treatment led to a profound inhibition of bone turnover in control mice, it significantly reduced osteoclast numbers in iron-treated mice without further impacting the bone formation rate or serum PINP levels. Our observations suggest that even though bone formation is markedly suppressed by iron overload, osteoclasts also play a key role in iron overload-induced bone loss and highlight them as potential therapeutic targets.
Collapse
Affiliation(s)
- Vanessa Passin
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Maria G. Ledesma‐Colunga
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Sandro Altamura
- Department of Pediatric Hematology, Oncology and ImmunologyUniversity of HeidelbergHeidelbergGermany
- Molecular Medicine Partnership UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Martina U. Muckenthaler
- Department of Pediatric Hematology, Oncology and ImmunologyUniversity of HeidelbergHeidelbergGermany
- Molecular Medicine Partnership UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Ulrike Baschant
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Lorenz C. Hofbauer
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy AgingMedical Faculty and University Hospital Carl Gustav Carus, Dresden University of TechnologyDresdenGermany
| |
Collapse
|
4
|
Hao Z, Guo X, Li L, Lei X, Tang Z, Zhai M, Yuan J. Identification of core genes and molecular prediction of drug targets for countering BPA-induced olfactory bulb neurotoxicity in male mice. Food Chem Toxicol 2024; 194:115098. [PMID: 39522797 DOI: 10.1016/j.fct.2024.115098] [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: 07/31/2024] [Revised: 09/28/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Bisphenol A (BPA) is ubiquitous in plastics, which can modify and improve the applicability and durability of plastics. Previous laboratory studies have shown that BPA can trigger cognitive impairment and depression. The olfactory bulb (OB) is significantly related to cognition and depression. However, there is a deficiency in information on BPA-induced OB neurotoxicity. Therefore, we analyzed the OB tissues of male mice at the transcriptional level after BPA poisoning at four different levels of concentration (0, 0.01, 0.1, and 1 μg/mL). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and weighted gene co-expression network analysis (WGCNA) were used to screen critical pathways and core genes. The result demonstrated that the PI3K-AKT signaling pathway might play a crucial role in the effects of BPA on the OB. In addition, two genes of the PI3K-AKT signaling pathway, the colony stimulating factor-1 receptor (Csf1r) and the toll-like receptor 2 (Tlr2), were screened by the protein-protein interaction networks. Furthermore, molecular docking identified ceftolozane as a potential drug candidate that could counteract BPA-related OB neurotoxicity. Conclusively, our results confirmed that BPA induced OB damage in male mice through the PI3K-AKT pathway and proposed that ceftolozane might reduce BPA-induced OB neurotoxicity.
Collapse
Affiliation(s)
- Zhoujie Hao
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Xin Guo
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Li Li
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China; Shanxi Key Laboratory of Ecological Animal Sciences and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Xuepei Lei
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Zhongwei Tang
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China; Shanxi Key Laboratory of Ecological Animal Sciences and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China
| | - Mengyu Zhai
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China
| | - Jianqin Yuan
- College of Life Sciences, Shanxi Agricultural University, Taigu, Shanxi 030801, China; Shanxi Key Laboratory of Ecological Animal Sciences and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| |
Collapse
|
5
|
Weinberger T, Denise M, Joppich M, Fischer M, Garcia Rodriguez C, Kumaraswami K, Wimmler V, Ablinger S, Räuber S, Fang J, Liu L, Liu WH, Winterhalter J, Lichti J, Thomas L, Esfandyari D, Percin G, Matin S, Hidalgo A, Waskow C, Engelhardt S, Todica A, Zimmer R, Pridans C, Gomez Perdiguero E, Schulz C. Resident and recruited macrophages differentially contribute to cardiac healing after myocardial ischemia. eLife 2024; 12:RP89377. [PMID: 38775664 PMCID: PMC11111219 DOI: 10.7554/elife.89377] [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] [Indexed: 05/24/2024] Open
Abstract
Cardiac macrophages are heterogenous in phenotype and functions, which has been associated with differences in their ontogeny. Despite extensive research, our understanding of the precise role of different subsets of macrophages in ischemia/reperfusion (I/R) injury remains incomplete. We here investigated macrophage lineages and ablated tissue macrophages in homeostasis and after I/R injury in a CSF1R-dependent manner. Genomic deletion of a fms-intronic regulatory element (FIRE) in the Csf1r locus resulted in specific absence of resident homeostatic and antigen-presenting macrophages, without affecting the recruitment of monocyte-derived macrophages to the infarcted heart. Specific absence of homeostatic, monocyte-independent macrophages altered the immune cell crosstalk in response to injury and induced proinflammatory neutrophil polarization, resulting in impaired cardiac remodeling without influencing infarct size. In contrast, continuous CSF1R inhibition led to depletion of both resident and recruited macrophage populations. This augmented adverse remodeling after I/R and led to an increased infarct size and deterioration of cardiac function. In summary, resident macrophages orchestrate inflammatory responses improving cardiac remodeling, while recruited macrophages determine infarct size after I/R injury. These findings attribute distinct beneficial effects to different macrophage populations in the context of myocardial infarction.
Collapse
Affiliation(s)
- Tobias Weinberger
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement et Cellules SouchesParisFrance
| | - Messerer Denise
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig Maximilian UniversityMunichGermany
| | - Maximilian Fischer
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
| | - Clarisabel Garcia Rodriguez
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement et Cellules SouchesParisFrance
| | - Konda Kumaraswami
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Vanessa Wimmler
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Sonja Ablinger
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Saskia Räuber
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- Department of Neurology, Medical Faculty, Heinrich Heine University of DüsseldorfDüsseldorfGermany
| | - Jiahui Fang
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Lulu Liu
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Wing Han Liu
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
| | - Julia Winterhalter
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
| | - Johannes Lichti
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
| | - Lukas Thomas
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
| | - Dena Esfandyari
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Institute of Pharmacology and Toxicology, Technical University MunichMunichGermany
| | - Guelce Percin
- Immunology of Aging, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI)JenaGermany
| | - Sandra Matin
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos IIIMadridSpain
| | - Andrés Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos IIIMadridSpain
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of MedicineNew HavenUnited States
| | - Claudia Waskow
- Immunology of Aging, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI)JenaGermany
- Faculty of Biological Sciences, Friedrich-Schiller-UniversityJenaGermany
| | - Stefan Engelhardt
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Institute of Pharmacology and Toxicology, Technical University MunichMunichGermany
| | - Andrei Todica
- Department of Nuclear Medicine, Ludwig Maximilian UniversityMunichGermany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig Maximilian UniversityMunichGermany
| | - Clare Pridans
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research InstituteEdinburghUnited Kingdom
| | - Elisa Gomez Perdiguero
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Département de Biologie du Développement et Cellules SouchesParisFrance
| | - Christian Schulz
- Medical Clinic I., Department of Cardiology, University Hospital, Ludwig Maximilian UniversityMunichGermany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine UniversityMunichGermany
- DZHK (German Centre for Cardiovascular Research), Partner site Munich Heart AllianceMunichGermany
- Department of Immunopharmacology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg UniversityMannheimGermany
| |
Collapse
|
6
|
Chen Z, Guo Y, Sun H, Zhang W, Hou S, Guo Y, Ma X, Meng H. Exploration of the causal associations between circulating inflammatory proteins, immune cells, and neuromyelitis optica spectrum disorder: a bidirectional Mendelian randomization study and mediation analysis. Front Aging Neurosci 2024; 16:1394738. [PMID: 38737586 PMCID: PMC11088236 DOI: 10.3389/fnagi.2024.1394738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Background An increasing body of research has demonstrated a robust correlation between circulating inflammatory proteins and neuromyelitis optica spectrum disorders (NMOSD). However, whether this association is causal or whether immune cells act as mediators currently remains unclear. Methods We employed bidirectional two-sample Mendelian randomization (TSMR) analysis to examine the potential causal association between circulating inflammatory proteins, immune cells, and NMOSD using data from genome-wide association studies (GWAS). Five different methods for Mendelian randomization analyses were applied, with the inverse variance-weighted (IVW) method being the primary approach. Sensitivity analyses were further performed to assess the presence of horizontal pleiotropy and heterogeneity in the results. Finally, a two-step Mendelian randomization (MR) design was employed to examine the potential mediating effects of immune cells. Results A notable causal relationship was observed between three circulating inflammatory proteins (CSF-1, IL-24, and TNFRSF9) and genetically predicted NMOSD. Furthermore, two immune cell phenotypes, genetically predicted CD8 on naive CD8+ T cells, and Hematopoietic Stem Cell Absolute Count were negatively and positively associated with genetically predicted NMOSD, respectively, although they did not appear to function as mediators. Conclusion Circulating inflammatory proteins and immune cells are causally associated with NMOSD. Immune cells do not appear to mediate the pathway linking circulating inflammatory proteins to NMOSD.
Collapse
Affiliation(s)
- Zhiqing Chen
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yujin Guo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Huaiyu Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Wuqiong Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Shuai Hou
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Guo
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaohui Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hongmei Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Bridges K, Pizzurro GA, Khunte M, Chen M, Salvador Rocha E, Alexander AF, Bass V, Kellman LN, Baskaran J, Miller-Jensen K. Single-Cell Analysis Reveals a Subset of High IL-12p40-Secreting Dendritic Cells within Mouse Bone Marrow-Derived Macrophages Differentiated with M-CSF. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1357-1365. [PMID: 38416039 DOI: 10.4049/jimmunol.2300431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024]
Abstract
Macrophages and dendritic cells (DCs), although ontogenetically distinct, have overlapping functions and exhibit substantial cell-to-cell heterogeneity that can complicate their identification and obscure innate immune function. In this study, we report that M-CSF-differentiated murine bone marrow-derived macrophages (BMDMs) exhibit extreme heterogeneity in the production of IL-12, a key proinflammatory cytokine linking innate and adaptive immunity. A microwell secretion assay revealed that a small fraction of BMDMs stimulated with LPS secrete most IL-12p40, and we confirmed that this is due to extremely high expression of Il12b, the gene encoding IL-12p40, in a subset of cells. Using an Il12b-YFP reporter mouse, we isolated cells with high LPS-induced Il12b expression and found that this subset was enriched for genes associated with the DC lineage. Single-cell RNA sequencing data confirmed a DC-like subset that differentiates within BMDM cultures that is transcriptionally distinct but could not be isolated by surface marker expression. Although not readily apparent in the resting state, upon LPS stimulation, this subset exhibited a typical DC-associated activation program that is distinct from LPS-induced stochastic BMDM cell-to-cell heterogeneity. Overall, our findings underscore the difficulty in distinguishing macrophages and DCs even in widely used in vitro murine BMDM cultures and could affect the interpretation of some studies that use BMDMs to explore acute inflammatory responses.
Collapse
Affiliation(s)
- Kate Bridges
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | | | - Mihir Khunte
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Meibin Chen
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | | | | | - Victor Bass
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT
| | - Laura N Kellman
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT
| | - Janani Baskaran
- Department of Biomedical Engineering, Yale University, New Haven, CT
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, CT
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT
| |
Collapse
|
8
|
Simkin J, Aloysius A, Adam M, Safaee F, Donahue RR, Biswas S, Lakhani Z, Gensel JC, Thybert D, Potter S, Seifert AW. Tissue-resident macrophages specifically express Lactotransferrin and Vegfc during ear pinna regeneration in spiny mice. Dev Cell 2024; 59:496-516.e6. [PMID: 38228141 PMCID: PMC10922778 DOI: 10.1016/j.devcel.2023.12.017] [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: 01/17/2022] [Revised: 05/30/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024]
Abstract
The details of how macrophages control different healing trajectories (regeneration vs. scar formation) remain poorly defined. Spiny mice (Acomys spp.) can regenerate external ear pinnae tissue, whereas lab mice (Mus musculus) form scar tissue in response to an identical injury. Here, we used this dual species system to dissect macrophage phenotypes between healing modes. We identified secreted factors from activated Acomys macrophages that induce a pro-regenerative phenotype in fibroblasts from both species. Transcriptional profiling of Acomys macrophages and subsequent in vitro tests identified VEGFC, PDGFA, and Lactotransferrin (LTF) as potential pro-regenerative modulators. Examining macrophages in vivo, we found that Acomys-resident macrophages secreted VEGFC and LTF, whereas Mus macrophages do not. Lastly, we demonstrate the requirement for VEGFC during regeneration and find that interrupting lymphangiogenesis delays blastema and new tissue formation. Together, our results demonstrate that cell-autonomous mechanisms govern how macrophages react to the same stimuli to differentially produce factors that facilitate regeneration.
Collapse
Affiliation(s)
- Jennifer Simkin
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA; Department of Orthopaedic Surgery, LSU Health-New Orleans, New Orleans, LA 70112, USA.
| | - Ajoy Aloysius
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Mike Adam
- Department of Pediatrics, University of Cincinnati Children's Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Fatemeh Safaee
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Renée R Donahue
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Shishir Biswas
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - Zohaib Lakhani
- Department of Orthopaedic Surgery, LSU Health-New Orleans, New Orleans, LA 70112, USA
| | - John C Gensel
- Department of Physiology, University of Kentucky, Lexington, KY 40506, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40506, USA
| | - David Thybert
- European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Steven Potter
- Department of Pediatrics, University of Cincinnati Children's Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
9
|
Ghasemi A, Martinez-Usatorre A, Li L, Hicham M, Guichard A, Marcone R, Fournier N, Torchia B, Martinez Bedoya D, Davanture S, Fernández-Vaquero M, Fan C, Janzen J, Mohammadzadeh Y, Genolet R, Mansouri N, Wenes M, Migliorini D, Heikenwalder M, De Palma M. Cytokine-armed dendritic cell progenitors for antigen-agnostic cancer immunotherapy. NATURE CANCER 2024; 5:240-261. [PMID: 37996514 PMCID: PMC10899110 DOI: 10.1038/s43018-023-00668-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 10/11/2023] [Indexed: 11/25/2023]
Abstract
Dendritic cells (DCs) are antigen-presenting myeloid cells that regulate T cell activation, trafficking and function. Monocyte-derived DCs pulsed with tumor antigens have been tested extensively for therapeutic vaccination in cancer, with mixed clinical results. Here, we present a cell-therapy platform based on mouse or human DC progenitors (DCPs) engineered to produce two immunostimulatory cytokines, IL-12 and FLT3L. Cytokine-armed DCPs differentiated into conventional type-I DCs (cDC1) and suppressed tumor growth, including melanoma and autochthonous liver models, without the need for antigen loading or myeloablative host conditioning. Tumor response involved synergy between IL-12 and FLT3L and was associated with natural killer and T cell infiltration and activation, M1-like macrophage programming and ischemic tumor necrosis. Antitumor immunity was dependent on endogenous cDC1 expansion and interferon-γ signaling but did not require CD8+ T cell cytotoxicity. Cytokine-armed DCPs synergized effectively with anti-GD2 chimeric-antigen receptor (CAR) T cells in eradicating intracranial gliomas in mice, illustrating their potential in combination therapies.
Collapse
Affiliation(s)
- Ali Ghasemi
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Amaia Martinez-Usatorre
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Luqing Li
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Mehdi Hicham
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Alan Guichard
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Rachel Marcone
- Agora Cancer Research Center, Lausanne, Switzerland
- Translational Data Science (TDS) Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Center, Lausanne, Switzerland
- Translational Data Science (TDS) Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Bruno Torchia
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Darel Martinez Bedoya
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Suzel Davanture
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mirian Fernández-Vaquero
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chaofan Fan
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Janzen
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yahya Mohammadzadeh
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Raphael Genolet
- Ludwig Institute for Cancer Research, Lausanne, Switzerland
- Department of Oncology, University of Lausanne (UNIL) and Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Nahal Mansouri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
| | - Mathias Wenes
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Denis Migliorini
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva (UNIGE), Geneva, Switzerland
- Department of Oncology, Geneva University Hospital (HUG), Geneva, Switzerland
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- The M3 Research Center, Eberhard Karls University, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180), Eberhard Karls University, Tübingen, Germany
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
| |
Collapse
|
10
|
Ding L, Wang Q, Martincuks A, Kearns MJ, Jiang T, Lin Z, Cheng X, Qian C, Xie S, Kim HJ, Launonen IM, Färkkilä A, Roberts TM, Freeman GJ, Liu JF, Konstantinopoulos PA, Matulonis U, Yu H, Zhao JJ. STING agonism overcomes STAT3-mediated immunosuppression and adaptive resistance to PARP inhibition in ovarian cancer. J Immunother Cancer 2023; 11:e005627. [PMID: 36609487 PMCID: PMC9827255 DOI: 10.1136/jitc-2022-005627] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Poly (ADP-ribose) polymerase (PARP) inhibition (PARPi) has demonstrated potent therapeutic efficacy in patients with BRCA-mutant ovarian cancer. However, acquired resistance to PARPi remains a major challenge in the clinic. METHODS PARPi-resistant ovarian cancer mouse models were generated by long-term treatment of olaparib in syngeneic Brca1-deficient ovarian tumors. Signal transducer and activator of transcription 3 (STAT3)-mediated immunosuppression was investigated in vitro by co-culture experiments and in vivo by analysis of immune cells in the tumor microenvironment (TME) of human and mouse PARPi-resistant tumors. Whole genome transcriptome analysis was performed to assess the antitumor immunomodulatory effect of STING (stimulator of interferon genes) agonists on myeloid cells in the TME of PARPi-resistant ovarian tumors. A STING agonist was used to overcome STAT3-mediated immunosuppression and acquired PARPi resistance in syngeneic and patient-derived xenografts models of ovarian cancer. RESULTS In this study, we uncover an adaptive resistance mechanism to PARP inhibition mediated by tumor-associated macrophages (TAMs) in the TME. Markedly increased populations of protumor macrophages are found in BRCA-deficient ovarian tumors that rendered resistance to PARPi in both murine models and patients. Mechanistically, PARP inhibition elevates the STAT3 signaling pathway in tumor cells, which in turn promotes protumor polarization of TAMs. STAT3 ablation in tumor cells mitigates polarization of protumor macrophages and increases tumor-infiltrating T cells on PARP inhibition. These findings are corroborated in patient-derived, PARPi-resistant BRCA1-mutant ovarian tumors. Importantly, STING agonists reshape the immunosuppressive TME by reprogramming myeloid cells and overcome the TME-dependent adaptive resistance to PARPi in ovarian cancer. This effect is further enhanced by addition of the programmed cell death protein-1 blockade. CONCLUSIONS We elucidate an adaptive immunosuppression mechanism rendering resistance to PARPi in BRCA1-mutant ovarian tumors. This is mediated by enrichment of protumor TAMs propelled by PARPi-induced STAT3 activation in tumor cells. We also provide a new strategy to reshape the immunosuppressive TME with STING agonists and overcome PARPi resistance in ovarian cancer.
Collapse
Affiliation(s)
- Liya Ding
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Qiwei Wang
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Antons Martincuks
- Immuno-Oncology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Michael J Kearns
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Tao Jiang
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ziying Lin
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Respiratory and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xin Cheng
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Changli Qian
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Shaozhen Xie
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hye-Jung Kim
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Anniina Färkkilä
- Obstetrics and Gynecology, University of Helsinki, Helsinki, Finland
| | - Thomas M Roberts
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Gordon J Freeman
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Joyce F Liu
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | | - Ursula Matulonis
- Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hua Yu
- Immuno-Oncology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Jean J Zhao
- Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
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: 42] [Impact Index Per Article: 21.0] [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
|
12
|
Wiechers C, Pezoldt J, Beckstette M, Berner J, Schraml BU, Huehn J. Lymph node stromal cells support the maturation of pre‐DCs into cDC‐like cells via colony‐stimulating factor 1. Immunology 2022; 166:475-491. [DOI: 10.1111/imm.13497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/18/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Carolin Wiechers
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
| | - Joern Pezoldt
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
- Laboratory of Systems Biology and Genetics, École Polytechnique Fédérale de Lausanne Lausanne Switzerland
| | - Michael Beckstette
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine Helmholtz Centre for Infection Research and Hannover Medical School Hannover Germany
| | - Johanna Berner
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich Planegg‐Martinsried Germany
- Walter‐Brendel‐Centre of Experimental Medicine University Hospital, LMU Munich Planegg‐Martinsried Germany
| | - Barbara U. Schraml
- Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, Faculty of Medicine, LMU Munich Planegg‐Martinsried Germany
- Walter‐Brendel‐Centre of Experimental Medicine University Hospital, LMU Munich Planegg‐Martinsried Germany
| | - Jochen Huehn
- Department Experimental Immunology Helmholtz Centre for Infection Research Braunschweig Germany
| |
Collapse
|
13
|
Hwang D, Seyedsadr MS, Ishikawa LLW, Boehm A, Sahin Z, Casella G, Jang S, Gonzalez MV, Garifallou JP, Hakonarson H, Zhang W, Xiao D, Rostami A, Zhang GX, Ciric B. CSF-1 maintains pathogenic but not homeostatic myeloid cells in the central nervous system during autoimmune neuroinflammation. Proc Natl Acad Sci U S A 2022; 119:e2111804119. [PMID: 35353625 PMCID: PMC9168454 DOI: 10.1073/pnas.2111804119] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 01/14/2022] [Indexed: 12/16/2022] Open
Abstract
The receptor for colony stimulating factor 1 (CSF-1R) is important for the survival and function of myeloid cells that mediate pathology during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). CSF-1 and IL-34, the ligands of CSF-1R, have similar bioactivities but distinct tissue and context-dependent expression patterns, suggesting that they have different roles. This could be the case in EAE, given that CSF-1 expression is up-regulated in the CNS, while IL-34 remains constitutively expressed. We found that targeting CSF-1 with neutralizing antibody halted ongoing EAE, with efficacy superior to CSF-1R inhibitor BLZ945, whereas IL-34 neutralization had no effect, suggesting that pathogenic myeloid cells were maintained by CSF-1. Both anti–CSF-1 and BLZ945 treatment greatly reduced the number of monocyte-derived cells and microglia in the CNS. However, anti–CSF-1 selectively depleted inflammatory microglia and monocytes in inflamed CNS areas, whereas BLZ945 depleted virtually all myeloid cells, including quiescent microglia, throughout the CNS. Anti–CSF-1 treatment reduced the size of demyelinated lesions and microglial activation in the gray matter. Lastly, we found that bone marrow–derived immune cells were the major mediators of CSF-1R–dependent pathology, while microglia played a lesser role. Our findings suggest that targeting CSF-1 could be effective in ameliorating MS pathology, while preserving the homeostatic functions of myeloid cells, thereby minimizing risks associated with ablation of CSF-1R–dependent cells.
Collapse
Affiliation(s)
- Daniel Hwang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Maryam S. Seyedsadr
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | | | - Alexandra Boehm
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ziver Sahin
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Giacomo Casella
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Soohwa Jang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Michael V. Gonzalez
- The Children’s Hospital of Philadelphia, Abramson Research Center, Center for Applied Genomics, Philadelphia, PA 19104
| | - James P. Garifallou
- The Children’s Hospital of Philadelphia, Abramson Research Center, Center for Applied Genomics, Philadelphia, PA 19104
| | - Hakon Hakonarson
- The Children’s Hospital of Philadelphia, Abramson Research Center, Center for Applied Genomics, Philadelphia, PA 19104
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Weifeng Zhang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Dan Xiao
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Abdolmohamad Rostami
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Guang-Xian Zhang
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| | - Bogoljub Ciric
- Department of Neurology, Jefferson Hospital for Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
14
|
Tomas L, Prica F, Schulz C. Trafficking of Mononuclear Phagocytes in Healthy Arteries and Atherosclerosis. Front Immunol 2021; 12:718432. [PMID: 34759917 PMCID: PMC8573388 DOI: 10.3389/fimmu.2021.718432] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Monocytes and macrophages play essential roles in all stages of atherosclerosis – from early precursor lesions to advanced stages of the disease. Intima-resident macrophages are among the first cells to be confronted with the influx and retention of apolipoprotein B-containing lipoproteins at the onset of hypercholesterolemia and atherosclerosis development. In this review, we outline the trafficking of monocytes and macrophages in and out of the healthy aorta, as well as the adaptation of their migratory behaviour during hypercholesterolemia. Furthermore, we discuss the functional and ontogenetic composition of the aortic pool of mononuclear phagocytes and its link to the atherosclerotic disease process. The development of mouse models of atherosclerosis regression in recent years, has enabled scientists to investigate the behaviour of monocytes and macrophages during the resolution of atherosclerosis. Herein, we describe the dynamics of these mononuclear phagocytes upon cessation of hypercholesterolemia and how they contribute to the restoration of tissue homeostasis. The aim of this review is to provide an insight into the trafficking, fate and disease-relevant dynamics of monocytes and macrophages during atherosclerosis, and to highlight remaining questions. We focus on the results of rodent studies, as analysis of cellular fates requires experimental manipulations that cannot be performed in humans but point out findings that could be replicated in human tissues. Understanding of the biology of macrophages in atherosclerosis provides an important basis for the development of therapeutic strategies to limit lesion formation and promote plaque regression.
Collapse
Affiliation(s)
- Lukas Tomas
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Filip Prica
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Schulz
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
15
|
Nobs SP, Kopf M. Tissue-resident macrophages: guardians of organ homeostasis. Trends Immunol 2021; 42:495-507. [PMID: 33972166 DOI: 10.1016/j.it.2021.04.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Tissue-resident macrophages (MTR) have recently emerged as a key rheostat capable of regulating the balance between organ health and disease. In most organs, ontogenetically and functionally distinct macrophage subsets fulfill a plethora of functions specific to their tissue environment. In this review, we summarize recent findings regarding the ontogeny and functions of macrophage populations in different mammalian tissues, describing how these cells regulate tissue homeostasis and how they can contribute to inflammation. Furthermore, we highlight new developments concerning certain general principles of tissue macrophage biology, including the importance of metabolism for understanding macrophage activation states and the influence of intrinsic and extrinsic factors on macrophage metabolic control. We also shed light on certain open questions in the field and how answering these might pave the way for tissue-specific therapeutic approaches.
Collapse
Affiliation(s)
- Samuel Philip Nobs
- Department of Immunology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
16
|
Neo WH, Lie-A-Ling M, Fadlullah MZH, Lacaud G. Contributions of Embryonic HSC-Independent Hematopoiesis to Organogenesis and the Adult Hematopoietic System. Front Cell Dev Biol 2021; 9:631699. [PMID: 33681211 PMCID: PMC7930747 DOI: 10.3389/fcell.2021.631699] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
During ontogeny, the establishment of the hematopoietic system takes place in several phases, separated both in time and location. The process is initiated extra-embryonically in the yolk sac (YS) and concludes in the main arteries of the embryo with the formation of hematopoietic stem cells (HSC). Initially, it was thought that HSC-independent hematopoietic YS cells were transient, and only required to bridge the gap to HSC activity. However, in recent years it has become clear that these cells also contribute to embryonic organogenesis, including the emergence of HSCs. Furthermore, some of these early HSC-independent YS cells persist into adulthood as distinct hematopoietic populations. These previously unrecognized abilities of embryonic HSC-independent hematopoietic cells constitute a new field of interest. Here, we aim to provide a succinct overview of the current knowledge regarding the contribution of YS-derived hematopoietic cells to the development of the embryo and the adult hematopoietic system.
Collapse
Affiliation(s)
- Wen Hao Neo
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| | - Michael Lie-A-Ling
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| | | | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Macclesfield, United Kingdom
| |
Collapse
|
17
|
Weinberger T, Esfandyari D, Messerer D, Percin G, Schleifer C, Thaler R, Liu L, Stremmel C, Schneider V, Vagnozzi RJ, Schwanenkamp J, Fischer M, Busch K, Klapproth K, Ishikawa-Ankerhold H, Klösges L, Titova A, Molkentin JD, Kobayashi Y, Engelhardt S, Massberg S, Waskow C, Perdiguero EG, Schulz C. Ontogeny of arterial macrophages defines their functions in homeostasis and inflammation. Nat Commun 2020; 11:4549. [PMID: 32917889 PMCID: PMC7486394 DOI: 10.1038/s41467-020-18287-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
Arterial macrophages have different developmental origins, but the association of macrophage ontogeny with their phenotypes and functions in adulthood is still unclear. Here, we combine macrophage fate-mapping analysis with single-cell RNA sequencing to establish their cellular identity during homeostasis, and in response to angiotensin-II (AngII)-induced arterial inflammation. Yolk sac erythro-myeloid progenitors (EMP) contribute substantially to adventitial macrophages and give rise to a defined cluster of resident immune cells with homeostatic functions that is stable in adult mice, but declines in numbers during ageing and is not replenished by bone marrow (BM)-derived macrophages. In response to AngII inflammation, increase in adventitial macrophages is driven by recruitment of BM monocytes, while EMP-derived macrophages proliferate locally and provide a distinct transcriptional response that is linked to tissue regeneration. Our findings thus contribute to the understanding of macrophage heterogeneity, and associate macrophage ontogeny with distinct functions in health and disease. Arterial macrophages develop from either yolk sac or bone marrow progenitors. Here, the author show that ageing-induced reduction of arterial macrophages is not replenished by bone marrow-derived cells, but under inflammatory conditions circulating monocytes are recruited to maintain homeostasis, while arterial macrophages of yolk sac origin carry out tissue repair.
Collapse
Affiliation(s)
- Tobias Weinberger
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Dena Esfandyari
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany.,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Strasse 29, 80802, Munich, Germany
| | - Denise Messerer
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Gulce Percin
- Regeneration in Hematopoiesis, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany
| | - Christian Schleifer
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Raffael Thaler
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Lulu Liu
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Christopher Stremmel
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Vanessa Schneider
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Ronald J Vagnozzi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Jennifer Schwanenkamp
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Maximilian Fischer
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Kay Klapproth
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Hellen Ishikawa-Ankerhold
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Lukas Klösges
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Anna Titova
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yasuhiro Kobayashi
- Institute for Oral Science, Matsumoto Dental University, 1780 Hiro-Oka Gobara Shiojiri, Nagano, 390-0781, Japan
| | - Stefan Engelhardt
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany.,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Strasse 29, 80802, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis, Leibniz-Institute on Aging - Fritz-Lipmann-Institute (FLI), Beutenbergstrasse 11, 07745, Jena, Germany.,Faculty of Biological Sciences, Friedrich-Schiller-University Jena, 07737 Jena, 07745, Jena, Germany
| | - Elisa Gomez Perdiguero
- Institut Pasteur, Macrophages and Endothelial cells, Département de Biologie du Développement et Cellules Souches, UMR3738 CNRS, Paris, 75015, France
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Marchioninistrasse 15, 81377, Munich, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802, Munich, Germany. .,Walter-Brendel-Center for Experimental Medicine, Ludwig Maximilian University, Marchioninistrasse 27, 81377, Munich, Germany.
| |
Collapse
|
18
|
Tian WT, Zhan FX, Liu Q, Luan XH, Zhang C, Shang L, Zhang BY, Pan SJ, Miao F, Hu J, Zhong P, Liu SH, Zhu ZY, Zhou HY, Sun S, Liu XL, Huang XJ, Jiang JW, Ma JF, Wang Y, Chen SF, Tang HD, Chen SD, Cao L. Clinicopathologic characterization and abnormal autophagy of CSF1R-related leukoencephalopathy. Transl Neurodegener 2019; 8:32. [PMID: 31827782 PMCID: PMC6886209 DOI: 10.1186/s40035-019-0171-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 08/21/2019] [Indexed: 01/13/2023] Open
Abstract
Background CSF1R-related leukoencephalopathy, also known as hereditary diffuse leukoencephalopathy with spheroids (HDLS), is a rare white-matter encephalopathy characterized by motor and neuropsychiatric symptoms due to colony-stimulating factor 1 receptor (CSF1R) gene mutation. Few of CSF1R mutations have been functionally testified and the pathogenesis remains unknown. Methods In order to investigate clinical and pathological characteristics of patients with CSF1R-related leukoencephalopathy and explore the potential impact of CSF1R mutations, we analyzed clinical manifestations of 15 patients from 10 unrelated families and performed brain biopsy in 2 cases. Next generation sequencing was conducted for 10 probands to confirm the diagnosis. Sanger sequencing, segregation analysis and phenotypic reevaluation were utilized to substantiate findings. Functional examination of identified mutations was further explored. Results Clinical and neuroimaging characteristics were summarized. The average age at onset was 35.9 ± 6.4 years (range 24–46 years old). Younger age of onset was observed in female than male (34.2 vs. 39.2 years). The most common initial symptoms were speech dysfunction, cognitive decline and parkinsonian symptoms. One patient also had marked peripheral neuropathy. Brain biopsy of two cases showed typical pathological changes, including myelin loss, axonal spheroids, phosphorylated neurofilament and activated macrophages. Electron microscopy disclosed increased mitochondrial vacuolation and disorganized neurofilaments in ballooned axons. A total of 7 pathogenic variants (4 novel, 3 documented) were identified with autophosphorylation deficiency, among which c.2342C > T remained partial function of autophosphorylation. Western blotting disclosed the significantly lower level of c.2026C > T (p.R676*) than wild type. The level of microtubule associated protein 1 light chain 3-II (LC3-II), a classical marker of autophagy, was significantly lower in mutants expressed cells than wild type group by western blotting and immunofluorescence staining. Conclusions Our findings support the loss-of-function and haploinsufficiency hypothesis in pathogenesis. Autophagy abnormality may play a role in the disease. Repairing or promoting the phosphorylation level of mutant CSF1R may shed light on therapeutic targets in the future. However, whether peripheral polyneuropathy potentially belongs to CSF1R-related spectrum deserves further study with longer follow-up and more patients enrolled. Trial registration ChiCTR, ChiCTR1800015295. Registered 21 March 2018. Electronic supplementary material The online version of this article (10.1186/s40035-019-0171-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wo-Tu Tian
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Fei-Xia Zhan
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Qing Liu
- 2Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100032 China
| | - Xing-Hua Luan
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Chao Zhang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China.,3Anhui University of Science and Technology School of Medicine, Huainan, 232001 Anhui Province China
| | - Liang Shang
- 2Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100032 China
| | - Ben-Yan Zhang
- 4Department of Pathology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Si-Jian Pan
- 5Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Fei Miao
- 6Department of Radiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jiong Hu
- 7Department of Hematology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Ping Zhong
- 8Suzhou Municipal Hospital, Suzhou, 234000 Anhui Province China
| | - Shi-Hua Liu
- 8Suzhou Municipal Hospital, Suzhou, 234000 Anhui Province China
| | - Ze-Yu Zhu
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hai-Yan Zhou
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Suya Sun
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiao-Li Liu
- 9Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406 China
| | - Xiao-Jun Huang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jing-Wen Jiang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jian-Fang Ma
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Ying Wang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Shu-Fen Chen
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Hui-Dong Tang
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Sheng-Di Chen
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Li Cao
- 1Department of Neurology, Rui Jin Hospital & Rui Jin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
19
|
Schwarzer P, Kokona D, Ebneter A, Zinkernagel MS. Effect of Inhibition of Colony-Stimulating Factor 1 Receptor on Choroidal Neovascularization in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 190:412-425. [PMID: 31783006 DOI: 10.1016/j.ajpath.2019.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 07/09/2019] [Accepted: 10/21/2019] [Indexed: 12/15/2022]
Abstract
Neovascular age-related macular degeneration is one of the leading causes of blindness. Microglia and macrophages play a critical role in choroidal neovascularization (CNV) and may, therefore, be potential targets to modulate the disease course. This study evaluated the effect of the colony-stimulating factor-1 receptor inhibitor PLX5622 on experimental laser-induced CNV. A 98% reduction of retinal microglia cells was observed in the retina 1 week after initiation of PLX5622 treatment, preventing accumulation of macrophages within the laser site and leading to a reduction of leukocytes within the choroid after CNV induction. Mice treated with PLX5622 had a significantly faster decrease of the CNV lesion size, as revealed by in vivo imaging and immunohistochemistry from day 3 to day 14 compared with untreated mice. Several inflammatory modulators, such as chemokine (C-C motif) ligand 9, granulocyte-macrophage colony-stimulating factor, soluble tumor necrosis factor receptor-I, IL-1α, and matrix metallopeptidase-2, were elevated in the acute phase of the disease when microglia were ablated with PLX5622, whereas other cytokines (eg, interferon-γ, IL-4, and IL-10) were reduced. Our results suggest that colony-stimulating factor-1 receptor inhibition may be a novel therapeutic target in patients with neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Petra Schwarzer
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Despina Kokona
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Andreas Ebneter
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern; and the Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
20
|
Jacome-Galarza CE, Percin GI, Muller JT, Mass E, Lazarov T, Eitler J, Rauner M, Yadav VK, Crozet L, Bohm M, Loyher PL, Karsenty G, Waskow C, Geissmann F. Developmental origin, functional maintenance and genetic rescue of osteoclasts. Nature 2019; 568:541-545. [PMID: 30971820 DOI: 10.1038/s41586-019-1105-7] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 03/06/2019] [Indexed: 11/09/2022]
Abstract
Osteoclasts are multinucleated giant cells that resorb bone, ensuring development and continuous remodelling of the skeleton and the bone marrow haematopoietic niche. Defective osteoclast activity leads to osteopetrosis and bone marrow failure1-9, whereas excess activity can contribute to bone loss and osteoporosis10. Osteopetrosis can be partially treated by bone marrow transplantation in humans and mice11-18, consistent with a haematopoietic origin of osteoclasts13,16,19 and studies that suggest that they develop by fusion of monocytic precursors derived from haematopoietic stem cells in the presence of CSF1 and RANK ligand1,20. However, the developmental origin and lifespan of osteoclasts, and the mechanisms that ensure maintenance of osteoclast function throughout life in vivo remain largely unexplored. Here we report that osteoclasts that colonize fetal ossification centres originate from embryonic erythro-myeloid progenitors21,22. These erythro-myeloid progenitor-derived osteoclasts are required for normal bone development and tooth eruption. Yet, timely transfusion of haematopoietic-stem-cell-derived monocytic cells in newborn mice is sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis. We also found that the postnatal maintenance of osteoclasts, bone mass and the bone marrow cavity involve iterative fusion of circulating blood monocytic cells with long-lived osteoclast syncytia. As a consequence, parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of haematopoietic-stem-cell chimerism, and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin K deficiency23,24. In sum, our results identify the developmental origin of osteoclasts and a mechanism that controls their maintenance in bones after birth. These data suggest strategies to rescue osteoclast deficiency in osteopetrosis and to modulate osteoclast activity in vivo.
Collapse
Affiliation(s)
- Christian E Jacome-Galarza
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gulce I Percin
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Dresden, Germany.,Regeneration in Hematopoiesis, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Faculty of Biological Sciences, Friedrich-Schiller University, Jena, Germany
| | - James T Muller
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elvira Mass
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Developmental Biology of the Innate Immune System, LIMES Institute, University of Bonn, Bonn, Germany
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jiri Eitler
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Faculty of Medicine, Dresden, Germany
| | - Vijay K Yadav
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Lucile Crozet
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mathieu Bohm
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre-Louis Loyher
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Claudia Waskow
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Dresden, Germany. .,Regeneration in Hematopoiesis, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Faculty of Biological Sciences, Friedrich-Schiller University, Jena, Germany. .,Department of Medicine III, Faculty of Medicine, Dresden, Germany.
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|